1
|
Nadora D, Ezzati S, Bol B, Aboud O. Serendipity in Neuro-Oncology: The Evolution of Chemotherapeutic Agents. Int J Mol Sci 2025; 26:2955. [PMID: 40243541 PMCID: PMC11988343 DOI: 10.3390/ijms26072955] [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: 01/08/2025] [Revised: 03/01/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
The development of novel therapeutics in neuro-oncology faces significant challenges, often marked by high costs and low success rates. Despite advances in molecular biology and genomics, targeted therapies have had limited impact on improving patient outcomes in brain tumors, particularly gliomas, due to the complex, multigenic nature of these malignancies. While significant efforts have been made to design drugs that target specific signaling pathways and genetic mutations, the clinical success of these rational approaches remains sparse. This review critically examines the landscape of neuro-oncology drug discovery, highlighting instances where serendipity has led to significant breakthroughs, such as the unexpected efficacy of repurposed drugs and off-target effects that proved beneficial. By exploring historical and contemporary cases, we underscore the role of chance in the discovery of impactful therapies, arguing that embracing serendipity alongside rational drug design may enhance future success in neuro-oncology drug development.
Collapse
Affiliation(s)
- Denise Nadora
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (S.E.); (B.B.)
| | - Shawyon Ezzati
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (S.E.); (B.B.)
| | - Brandon Bol
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (S.E.); (B.B.)
| | - Orwa Aboud
- Department of Neurology, Comprehensive Cancer Center, University of California, Davis, CA 95616, USA;
- Department of Neurological Surgery, Comprehensive Cancer Center, University of California, Davis, CA 95616, USA
| |
Collapse
|
2
|
Özbey G, Tuncay G, Düz SA, Çiğremiş Y, Karaer A. The Effect of Endometrial Polyp and Myoma Uteri on Fertility-Related Genes in the Endometrium. Reprod Sci 2025; 32:728-737. [PMID: 39915377 PMCID: PMC11870916 DOI: 10.1007/s43032-025-01802-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 01/22/2025] [Indexed: 03/03/2025]
Abstract
Endometrial polyps are hyperplastic overgrowths of the endometrium that contain both glands and stroma. Myoma uteri is the most common benign tumor of the female pelvis and uterus. HOXA10, which is involved during the organogenesis of the uterus in the embryonic period. The aim of this study was to compare the expression levels of infertility-related genes in endometrial tissue obtained from patients with endometrial polyp and myoma uteri and from healthy controls. A total of 36 patients, including 15 women with endometrial polyp and 21 women with myoma uteri, and 23 healthy controls were enrolled in the study. All patients were evaluated using transvaginal ultrasonography. Endometrial tissue samples were collected from the patient and control groups between the 19th and 21st days of the menstrual cycle. Expression levels of the receptivity markers PROK1, PROKR1, PROK2, PROKR2 and HOXA10 genes were determined by RT- PCR. When the patients diagnosed with endometrial polyp and the healthy controls were compared, it was observed statistically significantly that the expression of PROKR1 increased in endometrium tissue of patients with endometrial polyp (p < 0.05). In patients diagnosed with myoma uteri, gene expression levels of endometrial PROKR1 was statistically significant increased and gene expression levels of PROK1, PROKR2, HOXA10 were found to be statistically significantly decreased compared to the controls (p < 0.05). Changes in the endometrial expression of the HOXA10 and prokineticin gene family in patients with myoma uteri and endometrial polyps may explain certain aspects of infertility in these patients.
Collapse
Affiliation(s)
- Gürkan Özbey
- Department of Obstetrics and Gynecology, Faculty of Medicine, Adıyaman University, Adıyaman, Türkiye.
| | - Görkem Tuncay
- Department of Obstetrics and Gynecology, Faculty of Medicine, Inonu University, Malatya, Türkiye
| | - Senem Arda Düz
- Department of Obstetrics and Gynecology, Faculty of Medicine, Inonu University, Malatya, Türkiye
| | - Yılmaz Çiğremiş
- Department of Medical Biology and Genetics, Faculty of Medicine, Inonu University, Malatya, Türkiye
| | - Abdullah Karaer
- Department of Obstetrics and Gynecology, Faculty of Medicine, Inonu University, Malatya, Türkiye
| |
Collapse
|
3
|
Biswas N, Mori T, Ragava Chetty Nagaraj NK, Xin H, Diemer T, Li P, Su Y, Piermarocchi C, Ferrara N. Adenosine diphosphate stimulates VEGF-independent choroidal endothelial cell proliferation: A potential escape from anti-VEGF therapy. Proc Natl Acad Sci U S A 2025; 122:e2418752122. [PMID: 39835893 PMCID: PMC11789014 DOI: 10.1073/pnas.2418752122] [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: 09/13/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
We hypothesized that a strategy employing tissue-specific endothelial cells (EC) might facilitate the identification of tissue- or organ-specific vascular functions of ubiquitous metabolites. An unbiased approach was employed to identify water-soluble small molecules with mitogenic activity on choroidal EC. We identified adenosine diphosphate (ADP) as a candidate, following biochemical purification from mouse EL4 lymphoma extracts. ADP stimulated the growth of bovine choroidal EC (BCEC) and other bovine or human eye-derived EC. ADP induced rapid phosphorylation of extracellular signal-regulated kinase in a dose- and time-dependent manner. ADP-induced BCEC proliferation could be blocked by pretreatment with specific antagonists of the purinergic receptor P2Y1 but not with a vascular endothelial growth factor (VEGF) inhibitor, indicating that the EC mitogenic effects of ADP are not mediated by stimulation of the VEGF pathway. Intravitreal administration of ADP expanded the neovascular area in a mouse model of choroidal neovascularization. Single-cell transcriptomics from human choroidal datasets show the expression of P2RY1, but not other ADP receptors, in EC with a pattern similar to VEGFR2. Although ADP has been reported to be a growth inhibitor for vascular EC, here we describe its growth-stimulating effects for BCEC and other eye-derived EC.
Collapse
Affiliation(s)
- Nilima Biswas
- Department of Pathology, University of California San Diego, La Jolla, CA92093
| | - Tommaso Mori
- Department of Pathology, University of California San Diego, La Jolla, CA92093
| | | | - Hong Xin
- Department of Pathology, University of California San Diego, La Jolla, CA92093
| | - Tanja Diemer
- Department of Pathology, University of California San Diego, La Jolla, CA92093
| | - Pin Li
- Department of Pathology, University of California San Diego, La Jolla, CA92093
| | - Yongxuan Su
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA92093
| | - Carlo Piermarocchi
- Department of Physics and Astronomy, Michigan State University, East Lansing, MI48824
| | - Napoleone Ferrara
- Department of Pathology, University of California San Diego, La Jolla, CA92093
- Department of Ophthalmology, University of California San Diego, La Jolla, CA92093
| |
Collapse
|
4
|
Gomez-Salinero JM, Redmond D, Rafii S. Microenvironmental determinants of endothelial cell heterogeneity. Nat Rev Mol Cell Biol 2025:10.1038/s41580-024-00825-w. [PMID: 39875728 DOI: 10.1038/s41580-024-00825-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
During development, endothelial cells (ECs) undergo an extraordinary specialization by which generic capillary microcirculatory networks spanning from arteries to veins transform into patterned organotypic zonated blood vessels. These capillary ECs become specialized to support the cellular and metabolic demands of each specific organ, including supplying tissue-specific angiocrine factors that orchestrate organ development, maintenance of organ-specific functions and regeneration of injured adult organs. Here, we illustrate the mechanisms by which microenvironmental signals emanating from non-vascular niche cells induce generic ECs to acquire specific inter-organ and intra-organ functional attributes. We describe how perivascular, parenchymal and immune cells dictate vascular heterogeneity and capillary zonation, and how this system is maintained through tissue-specific signalling activated by vasculogenic and angiogenic factors and deposition of matrix components. We also discuss how perturbation of organotypic vascular niche cues lead to erasure of EC signatures, contributing to the pathogenesis of disease processes. We also describe approaches that use reconstitution of tissue-specific signatures of ECs to promote regeneration of damaged organs.
Collapse
Affiliation(s)
- Jesus M Gomez-Salinero
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
5
|
Sergent F, Vaiman D, Raia‐Barjat T, Younes H, Marquette C, Desseux M, Nahed RA, Kieu T, Dung NV, Keck M, Hoffmann P, Murthi P, Benharouga M, Alfaidy N. Antagonisation of Prokineticin Receptor-2 Attenuates Preeclampsia Symptoms. J Cell Mol Med 2025; 29:e70346. [PMID: 39817714 PMCID: PMC11736873 DOI: 10.1111/jcmm.70346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/18/2025] Open
Abstract
Preeclampsia (PE) is the most threatening pathology of human pregnancy. Placenta from PE patients releases harmful factors that contribute to the exacerbation of the disease. Among these factors is the prokineticin1 (PROK1) and its receptor, PROKR2 that we identified as a mediators of PE. Here we tested the effects of PKRA, an antagonist of PROKR2, on the attenuation of PE symptoms. We used the genetic PE mouse model, STOX1 that overexpresses Stox1 gene in a heterozygosis manner in the placenta. This model allowed exploiting two genotypes of the offspring, those that overexpress the Stox1 gene, and the WT that grow in a PE environment (STE). We characterised the effect PKRA (1 μM) on the attenuation of PE symptoms and compared its effects on STOX1 and STE placentas. We also used STOX1 overexpressing trophoblast cells to decipher the PROK1-underlying mechanism. We demonstrated that (i) antagonisation of PROKR2 attenuated PE-mediated hypertension and proteinuria, (ii) STE placentas and foetuses exhibited better outcomes in response to PKRA, (iii) the secretome of STOX1-trophoblasts impacted the integrity of the fetal vasculature that was attenuated by PKRA treatment. This study demonstrates the direct involvement of the PROK1 in PE and identifies PKRA as a promising therapy for PE.
Collapse
Affiliation(s)
- Frédéric Sergent
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Daniel Vaiman
- Institute Cochin, U1016, INSERM, UMR 8504 CNRS, Paris‐Descartes UniversitéParisFrance
| | - Tiphaine Raia‐Barjat
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Hadi Younes
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Christel Marquette
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Morgane Desseux
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Roland Abi Nahed
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Trinh‐Le‐Vi Kieu
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Nguyen Viet Dung
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Mathilde Keck
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies Pour la Santé (DMTS)Gif‐sur‐YvetteFrance
| | - Pascale Hoffmann
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
- Centre Hospitalo‐Universitaire Grenoble Alpes, Service Obstétrique, CS 10217GrenobleFrance
- Université Grenoble AlpesGrenobleFrance
| | - Padma Murthi
- Department of PharmacologyMonash Biomedicine Discovery Institute, Monash UniversityMelbourneVictoriaAustralia
- Department of Maternal‐Fetal Medicine Pregnancy Research CentreThe Royal Women's HospitalMelbourneVictoriaAustralia
- Department of Obstetrics and GynecologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Mohamed Benharouga
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Nadia Alfaidy
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
- Centre Hospitalo‐Universitaire Grenoble Alpes, Service Obstétrique, CS 10217GrenobleFrance
- Université Grenoble AlpesGrenobleFrance
| |
Collapse
|
6
|
Baryla M, Kaczynski P, Goryszewska-Szczurek E, Waclawik A. The regulation of the expression of prokineticin 1 and its receptors and its mechanism of action in the porcine corpus luteum. Theriogenology 2024; 226:39-48. [PMID: 38838613 DOI: 10.1016/j.theriogenology.2024.05.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
Prokineticin 1 (PROK1) is an important factor in pregnancy establishment in pigs, acting at the embryo-maternal interface and the corpus luteum (CL). Estradiol-17β (E2) is the primary pregnancy recognition signal in pigs, and its effects are augmented by luteotropic prostaglandin E2 (PGE2). On the contrary, prostaglandin F2α (PGF2α) exerts mainly a luteolytic effect. The present study aimed to elucidate whether E2, PGE2, and PGF2α regulate the expression of PROK1 and its receptors in the porcine CL and to determine the PROK1 effect on luteal endothelial cells and pathways that may be involved in this regulation. The effects of E2, PGE2, and PGF2α on the expressions of PROK1 and its receptors in the CL were studied using an in vitro model of ultrathin luteal tissue explants model. Additionally, the effects of E2 and PGE2 on the PROK1 system were determined using an in vivo approach, in which the hormones were administered into the uterine lumen to imitate their secretion by embryos. Endothelial cell proliferation was measured using the colorimetric method. E2 acting via estrogen receptors simulated the mRNA and protein expressions of PROK1 and PROKR1 in CL explants in vitro (p < 0.05). The simultaneous action of E2 with PGE2 enhanced the expression of luteal PROK1 mRNA in vitro (p < 0.05). Estradiol-17β acting alone significantly increased PROK1 mRNA levels in vivo, whereas E2 simultaneously administered with PGE2 significantly elevated the PROK1 mRNA expression and PROKR1 mRNA and protein contents in CLs adjacent to uterine horns receiving hormonal infusion compared with CLs adjacent to placebo-treated uterine horns (p < 0.01). The PROK1 protein expression was significantly higher in the CLs of pigs treated with E2, PGE2, and E2 together with PGE2 than in the control group. PGF2α increased the PROK1 mRNA content in CLs on days 12 and 14 of the estrous cycle (p < 0.05). The expression of PROKR2 at the mRNA and protein levels remained unchanged in response to in vitro and in vivo treatments. PROK1 stimulated the proliferation of luteal endothelial cells by activating the MAPK, AKT, and mTOR pathways (p < 0.05). In summary, the luteal expressions of PROK1 and PROKR1 in early pregnancy are regulated by E2 and PGE2. PROK1 stimulates luteal angiogenesis by activating the MAPK, AKT, and mTOR pathways. The regulation of luteal PROK1 expression by PGF2α indicates PROK1's putative role during luteolysis. We conclude that PROK1-PROKR1 signaling supports luteal function during CL rescue in pregnancy in pigs.
Collapse
Affiliation(s)
- Monika Baryla
- Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Piotr Kaczynski
- Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Ewelina Goryszewska-Szczurek
- Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Agnieszka Waclawik
- Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| |
Collapse
|
7
|
Ou KL, Chen CK, Huang JJ, Chang WW, Hsieh Li SM, Jiang TX, Widelitz RB, Lansford R, Chuong CM. Adaptive patterning of vascular network during avian skin development: Mesenchymal plasticity and dermal vasculogenesis. Cells Dev 2024; 179:203922. [PMID: 38688358 PMCID: PMC11633821 DOI: 10.1016/j.cdev.2024.203922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024]
Abstract
A vasculature network supplies blood to feather buds in the developing skin. Does the vasculature network during early skin development form by sequential sprouting from the central vasculature or does local vasculogenesis occur first that then connect with the central vascular tree? Using transgenic Japanese quail Tg(TIE1p.H2B-eYFP), we observe that vascular progenitor cells appear after feather primordia formation. The vasculature then radiates out from each bud and connects with primordial vessels from neighboring buds. Later they connect with the central vasculature. Epithelial-mesenchymal recombination shows local vasculature is patterned by the epithelium, which expresses FGF2 and VEGF. Perturbing noggin expression leads to abnormal vascularization. To study endothelial origin, we compare transcriptomes of TIE1p.H2B-eYFP+ cells collected from the skin and aorta. Endothelial cells from the skin more closely resemble skin dermal cells than those from the aorta. The results show developing chicken skin vasculature is assembled by (1) physiological vasculogenesis from the peripheral tissue, and (2) subsequently connects with the central vasculature. The work implies mesenchymal plasticity and convergent differentiation play significant roles in development, and such processes may be re-activated during adult regeneration. SUMMARY STATEMENT: We show the vasculature network in the chicken skin is assembled using existing feather buds as the template, and endothelia are derived from local bud dermis and central vasculature.
Collapse
Affiliation(s)
- Kuang-Ling Ou
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States of America; Ostrow School of Dentistry of the University of Southern California, Los Angeles, CA, United States of America; Burn Center, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Kuan Chen
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States of America
| | - Junxiang J Huang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, 1501 San Pablo Street, Los Angeles, CA, United States of America; Graduate Programs in Biomedical and Biological Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - William Weijen Chang
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States of America; Integrative Stem Cell Center, China Medical University, Taichung, Taiwan; Institute of Physiology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Shu-Man Hsieh Li
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States of America; Ostrow School of Dentistry of the University of Southern California, Los Angeles, CA, United States of America
| | - Ting-Xin Jiang
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States of America
| | - Randall B Widelitz
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States of America
| | - Rusty Lansford
- Department of Radiology and Developmental Neuroscience Program, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States of America; Department of Radiology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States of America
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States of America.
| |
Collapse
|
8
|
Perez-Gutierrez L, Li P, Ferrara N. Endothelial cell diversity: the many facets of the crystal. FEBS J 2024; 291:3287-3302. [PMID: 36266750 DOI: 10.1111/febs.16660] [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: 07/17/2022] [Revised: 10/03/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Endothelial cells (ECs) form the inner lining of blood vessels and play crucial roles in angiogenesis. While it has been known for a long time that there are considerable differences among ECs from lymphatic and blood vessels, as well as among arteries, veins and capillaries, the full repertoire of endothelial diversity is only beginning to be elucidated. It has become apparent that the role of ECs is not just limited to their exchange functions. Indeed, a multitude of organ-specific functions, including release of growth factors, regulation of immune functions, have been linked to ECs. Recent years have seen a surge into the identification of spatiotemporal molecular and functional heterogeneity of ECs, supported by technologies such as single-cell RNA sequencing (scRNA-seq), lineage tracing and intersectional genetics. Together, these techniques have spurred the generation of epigenomic, transcriptomic and proteomic signatures of ECs. It is now clear that ECs across organs and in different vascular beds, but even within the same vessel, have unique molecular identities and employ specialized molecular mechanisms to fulfil highly specialized needs. Here, we focus on the molecular heterogeneity of the endothelium in different organs and pathological conditions.
Collapse
Affiliation(s)
- Lorena Perez-Gutierrez
- Department of Pathology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Pin Li
- Department of Pathology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Napoleone Ferrara
- Department of Pathology, Moores Cancer Center, University of California, San Diego, CA, USA
| |
Collapse
|
9
|
Sinisi AA, Rossi V, Martino MD, Esposito F, Chieffi P. The role of endocrine gland derived vascular growth factor/ Prokineticin-1 in human prostate cells. GHM OPEN 2024; 4:37-41. [PMID: 40144745 PMCID: PMC11933967 DOI: 10.35772/ghmo.2023.01021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/07/2023] [Accepted: 12/26/2023] [Indexed: 03/28/2025]
Abstract
In steroidogenic tissues, a novel class of angiogenic molecules known as endocrine gland-derived vascular endothelial growth factors (EG-VEGF)/prokineticins are primarily produced. Here, we investigated how EG-VEGF/ PROK1, a member of PROKs family, and its receptor are able to affect cellular motility in both non-neoplastic and cancerous human prostate cells. Using Western blot and motility test studies, EPN cells, a non-transformed cell line and Cancer Epithelial Prostatic Cells (CEPC) were employed as cellular models in the current investigation. Western blot examination of EPN normal prostate cells treated with EG-VEGF/PROK1 revealed that ERK1/2 was rapidly phosphorylated within 5, 10, and 20 minutes, while CEPC had high and sustained ERK1/2 activity at the same periods. Then, compared to normal EPN prostate cells, CEPC treated with EG-VEGF/PROK1 for up to 72 hours demonstrated enhanced cell motility. Based on our findings, EG-VEGF/PROK1 may play a role in prostate cancer progression by controlling angiogenesis and the motility of metastatic cells in CEPC cells, likely as a consequence of ERK1/2 activation, as contrasted to EPN normal prostate cells.
Collapse
Affiliation(s)
- Antonio Agostino Sinisi
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Valentina Rossi
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Marco De Martino
- Institute of Endocrinology and Experimental Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Esposito
- Institute of Endocrinology and Experimental Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy
| | - Paolo Chieffi
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
10
|
Zhi S, Chen C, Huang H, Zhang Z, Zeng F, Zhang S. Hypoxia-inducible factor in breast cancer: role and target for breast cancer treatment. Front Immunol 2024; 15:1370800. [PMID: 38799423 PMCID: PMC11116789 DOI: 10.3389/fimmu.2024.1370800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Globally, breast cancer stands as the most prevalent form of cancer among women. The tumor microenvironment of breast cancer often exhibits hypoxia. Hypoxia-inducible factor 1-alpha, a transcription factor, is found to be overexpressed and activated in breast cancer, playing a pivotal role in the anoxic microenvironment by mediating a series of reactions. Hypoxia-inducible factor 1-alpha is involved in regulating downstream pathways and target genes, which are crucial in hypoxic conditions, including glycolysis, angiogenesis, and metastasis. These processes significantly contribute to breast cancer progression by managing cancer-related activities linked to tumor invasion, metastasis, immune evasion, and drug resistance, resulting in poor prognosis for patients. Consequently, there is a significant interest in Hypoxia-inducible factor 1-alpha as a potential target for cancer therapy. Presently, research on drugs targeting Hypoxia-inducible factor 1-alpha is predominantly in the preclinical phase, highlighting the need for an in-depth understanding of HIF-1α and its regulatory pathway. It is anticipated that the future will see the introduction of effective HIF-1α inhibitors into clinical trials, offering new hope for breast cancer patients. Therefore, this review focuses on the structure and function of HIF-1α, its role in advancing breast cancer, and strategies to combat HIF-1α-dependent drug resistance, underlining its therapeutic potential.
Collapse
Affiliation(s)
| | | | | | | | - Fancai Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Shujun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
11
|
Jeon HR, Kang JI, Bhang SH, Park KM, Kim DI. Transplantation of Stem Cell Spheroid-Laden 3-Dimensional Patches with Bioadhesives for the Treatment of Myocardial Infarction. Biomater Res 2024; 28:0007. [PMID: 38439926 PMCID: PMC10911933 DOI: 10.34133/bmr.0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/03/2024] [Indexed: 03/06/2024] Open
Abstract
Myocardial infarction (MI) is treated with stem cell transplantation using various biomaterials and methods, such as stem cell/spheroid injections, cell sheets, and cardiac patches. However, current treatment methods have some limitations, including low stem cell engraftment and poor therapeutic effects. Furthermore, these methods cause secondary damage to heart due to injection and suturing to immobilize them in the heart, inducing side effects. In this study, we developed stem cell spheroid-laden 3-dimensional (3D) patches (S_3DP) with biosealant to treat MI. This 3D patch has dual modules, such as open pockets to directly deliver the spheroids with their paracrine effects and closed pockets to improve the engraft rate by protecting the spheroid from harsh microenvironments. The spheroids formed within S_3DP showed increased viability and expression of angiogenic factors compared to 2-dimensional cultured cells. We also fabricated gelatin-based tissue adhesive biosealants via a thiol-ene reaction and disulfide bond formation. This biosealant showed stronger tissue adhesiveness than commercial fibrin glue. Furthermore, we successfully applied S_3DP using a biosealant in a rat MI model without suturing in vivo, thereby improving cardiac function and reducing heart fibrosis. In summary, S_3DP and biosealant have excellent potential as advanced stem cell therapies with a sutureless approach to MI treatment.
Collapse
Affiliation(s)
- Hye Ran Jeon
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST),
Sungkyunkwan University, Seoul 06355, Republic of Korea
| | - Jeon Il Kang
- Department of Bioengineering and Nano-Bioengineering, College of Life Sciences and Bioengineering,
Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering,
Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Kyung Min Park
- Department of Bioengineering and Nano-Bioengineering, College of Life Sciences and Bioengineering,
Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea
- Research Center for Bio Materials & Process Development,
Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea
| | - Dong-Ik Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST),
Sungkyunkwan University, Seoul 06355, Republic of Korea
- Division of Vascular Surgery,
Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea
| |
Collapse
|
12
|
Guo C, Cui Y, Jiao M, Yao J, Zhao J, Tian Y, Dong J, Liao L. Crosstalk between proximal tubular epithelial cells and other interstitial cells in tubulointerstitial fibrosis after renal injury. Front Endocrinol (Lausanne) 2024; 14:1256375. [PMID: 38260142 PMCID: PMC10801024 DOI: 10.3389/fendo.2023.1256375] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/22/2023] [Indexed: 01/24/2024] Open
Abstract
The energy needs of tubular epithelial components, especially proximal tubular epithelial cells (PTECs), are high and they heavily depend on aerobic metabolism. As a result, they are particularly vulnerable to various injuries caused by factors such as ischemia, proteinuria, toxins, and elevated glucose levels. Initial metabolic and phenotypic changes in PTECs after injury are likely an attempt at survival and repair. Nevertheless, in cases of recurrent or prolonged injury, PTECs have the potential to undergo a transition to a secretory state, leading to the generation and discharge of diverse bioactive substances, including transforming growth factor-β, Wnt ligands, hepatocyte growth factor, interleukin (IL)-1β, lactic acid, exosomes, and extracellular vesicles. By promoting fibroblast activation, macrophage recruitment, and endothelial cell loss, these bioactive compounds stimulate communication between epithelial cells and other interstitial cells, ultimately worsening renal damage. This review provides a summary of the latest findings on bioactive compounds that facilitate the communication between these cellular categories, ultimately leading to the advancement of tubulointerstitial fibrosis (TIF).
Collapse
Affiliation(s)
- Congcong Guo
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuying Cui
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- First Clinical Medical College, Shandong University of Traditional Chinese Medicin, Jinan, Shandong, China
| | - Mingwen Jiao
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Jinming Yao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Junyu Zhao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yutian Tian
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Jianjun Dong
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lin Liao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- First Clinical Medical College, Shandong University of Traditional Chinese Medicin, Jinan, Shandong, China
| |
Collapse
|
13
|
Kono H, Goi T, Matsunaka T, Koneri K. Anti-Prokineticin1 Suppresses Liver Metastatic Tumors in a Mouse Model of Colorectal Cancer with Liver Metastasis. Curr Issues Mol Biol 2023; 46:44-52. [PMID: 38275664 PMCID: PMC10814645 DOI: 10.3390/cimb46010004] [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: 10/30/2023] [Revised: 12/04/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
Multidisciplinary treatment for colorectal cancer (CRC) has undergone significant advances, and molecularly targeted drugs have substantially improved patient prognosis. However, one problem with current molecularly targeted therapeutics is that they must be used in combination with anticancer agents. New molecular targeted therapies that can be used alone are needed. We have previously identified prokineticin1 (PROK1) factor as a therapeutic potential target for CRC. PROK1 factor is involved in the angiogenesis of tissues surrounding CRC tumors. Additionally, PROK1 receptors 1 and 2 are expressed in CRC cell lines, playing roles in cell proliferation via an autocrine mechanism and in the signaling system. In this study, a liver metastasis mouse model was developed using human colorectal cancer cell lines, and mice were divided into anti-PROK1 antibody administration and control groups. Mice were treated intraperitoneally with antibodies or phosphate-buffered saline (control) every three days. The number, size, and cell proliferation ability of metastatic lesions were analyzed. Our results suggested that the number, size, and cancer cell proliferation ability of metastatic lesions decreased, and the survival time significantly increased in the antibody-treated group compared to those in the control group. Thus, the anti-PROK1 antibody therapy suppressed the cell proliferation ability of liver metastatic lesions in a CRC mouse model, suggesting its potential as a novel treatment strategy.
Collapse
Affiliation(s)
| | - Takanori Goi
- First Department of Surgery, University of Fukui, 23-3 Eiheiji-cho, Yoshida-gun, Fukui 9101193, Japan; (H.K.); (T.M.); (K.K.)
| | | | | |
Collapse
|
14
|
Goto S, Ozaki Y, Ozawa F, Yoshihara H, Ujvari D, Kitaori T, Sugiura-Ogasawara M. Impaired decidualization and relative increase of PROK1 expression in the decidua of patients with unexplained recurrent pregnancy loss showing insulin resistance. J Reprod Immunol 2023; 160:104155. [PMID: 37801889 DOI: 10.1016/j.jri.2023.104155] [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: 06/24/2023] [Revised: 08/27/2023] [Accepted: 09/20/2023] [Indexed: 10/08/2023]
Abstract
A recent meta-analysis revealed that patients with unexplained recurrent pregnancy loss (RPL) show higher insulin resistance compared to healthy controls. However, the etiology of RPL remains unknown. Prokineticin (PROK1), a pleiotropic uterine endometrial protein, is important for implantation and decidualization and is regulated by hypoxia and insulin. In this study, we investigated the decidualization status and the role of PROK1 in the decidua of patients with unexplained RPL showing insulin resistance. Thirty-two patients with unexplained RPL were included in this study. Following the diagnosis of a miscarriage, the decidua and villi of the patient were surgically collected. Fasting blood glucose and insulin levels were measured, and HOMA-β was calculated. Using IHC and ELISA, the expression of IGFBP-1, PRL and PROK1 in the decidua and IGF-2 in the villi were analyzed in patients with euploid miscarriage with a high HOMA-β index (n = 8) and compared to controls (euploid miscarriage with normal HOMA-β: n = 12, aneuploid miscarriage with normal HOMA-β: n = 12). The co-localization of PROK1 and IGFBP-1 was observed in the decidua by IHC. In the decidua of RPL patients with high HOMA-β, the expression levels of IGFBP-1 and PRL were significantly lower, whereas the PROK1/IGFBP-1 ratio was significantly higher compared to that of the controls. IGF-2 expression in villi was significantly lower in RPL patients with high HOMA-β. Impaired decidualization and excessive PROK1 production may have pathological implications in patients with unexplained RPL with insulin resistance, especially under the state of hyper insulin production.
Collapse
Affiliation(s)
- Shinobu Goto
- Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya, Aichi ZIP 467-8601 , Japan.
| | - Yasuhiko Ozaki
- Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya, Aichi ZIP 467-8601 , Japan; Department of Obstetrics and Gynecology, Nagoya City University West Medical Center, 1-1-1 Hirate-cho, Kita-ku, Nagoya, Aichi ZIP 462-8508, Japan
| | - Fumiko Ozawa
- Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya, Aichi ZIP 467-8601 , Japan
| | - Hiroyuki Yoshihara
- Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya, Aichi ZIP 467-8601 , Japan; Department of Microbiology, Tumor and Cell Biology, National Pandemic Centre, Centre for Translational Microbiome Research, Karolinska Institute, Solna, Sweden
| | - Dorina Ujvari
- Department of Women's and Children's Health, Karolinska Institute, Solna, Sweden; Division of Biomedical Sciences, Clinical Science Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Tamao Kitaori
- Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya, Aichi ZIP 467-8601 , Japan
| | - Mayumi Sugiura-Ogasawara
- Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-ku, Nagoya, Aichi ZIP 467-8601 , Japan
| |
Collapse
|
15
|
Raia-Barjat T, Chauleur C, Collet C, Rancon F, Hoffmann P, Desseux M, Lemaitre N, Benharouga M, Giraud A, Alfaidy N. EG-VEGF maternal levels predict spontaneous preterm birth in the second and third trimesters in pregnant women with risk factors for placenta-mediated complications. Sci Rep 2023; 13:19921. [PMID: 37963927 PMCID: PMC10645734 DOI: 10.1038/s41598-023-46883-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 11/06/2023] [Indexed: 11/16/2023] Open
Abstract
Prediction of spontaneous preterm birth in asymptomatic women remains a great challenge for the public health system. The aim of the study was to determine the informational value of EG-VEGF circulating levels for prediction of spontaneous preterm birth in the second and third trimesters in pregnant women at high risk for placenta-mediated complications. A prospective multicenter cohort study including 200 pregnant patients with five-serum sampling per patient. Women with spontaneous preterm birth have higher concentrations of serum EG-VEGF than uncomplicated patients at 24 weeks, 28 weeks and 32 weeks (p = 0.03, 0.02 and < 0.001). The areas under the curve reached 0.9 with 100% sensitivity at 32 weeks for the prediction of spontaneous preterm birth. Serum EG-VEGF concentrations could be considered as a reliable biomarker of spontaneous preterm birth in high-risk for placenta-mediated complications pregnant women.
Collapse
Affiliation(s)
- Tiphaine Raia-Barjat
- Department of Gynecology and Obstetrics, Hôpital Nord, University Hospital, Centre Hospitalier Universitaire de Saint-Étienne, Avenue Albert Raimond, 42270, Saint-Étienne, Saint Priest en Jarez, France.
- INSERM U1059 SAINBIOSE, Université Jean Monnet, Saint-Étienne, France.
- Institut National de La Santé et de la Recherche Médicale (INSERM), U1292, MAB2 Team, Laboratoire de BioSanté, Bât C3, Pièce 304B.17 rue des Martyrs, 38054, Grenoble, France.
| | - Céline Chauleur
- Department of Gynecology and Obstetrics, Hôpital Nord, University Hospital, Centre Hospitalier Universitaire de Saint-Étienne, Avenue Albert Raimond, 42270, Saint-Étienne, Saint Priest en Jarez, France
- INSERM U1059 SAINBIOSE, Université Jean Monnet, Saint-Étienne, France
| | - Constance Collet
- Institut National de La Santé et de la Recherche Médicale (INSERM), U1292, MAB2 Team, Laboratoire de BioSanté, Bât C3, Pièce 304B.17 rue des Martyrs, 38054, Grenoble, France
- Commissariat à l'Energie Atomique (CEA), DSV-IRIG, Grenoble, France
- Université Grenoble Alpes (UGA), Grenoble, France
| | - Florence Rancon
- INSERM U1059 SAINBIOSE, Université Jean Monnet, Saint-Étienne, France
- INSERM, Centre d'Investigation Clinique 1408, Saint-Étienne, France
| | - Pascale Hoffmann
- Institut National de La Santé et de la Recherche Médicale (INSERM), U1292, MAB2 Team, Laboratoire de BioSanté, Bât C3, Pièce 304B.17 rue des Martyrs, 38054, Grenoble, France
- Commissariat à l'Energie Atomique (CEA), DSV-IRIG, Grenoble, France
- Université Grenoble Alpes (UGA), Grenoble, France
| | - Morgane Desseux
- Institut National de La Santé et de la Recherche Médicale (INSERM), U1292, MAB2 Team, Laboratoire de BioSanté, Bât C3, Pièce 304B.17 rue des Martyrs, 38054, Grenoble, France
- Commissariat à l'Energie Atomique (CEA), DSV-IRIG, Grenoble, France
- Université Grenoble Alpes (UGA), Grenoble, France
| | - Nicolas Lemaitre
- Institut National de La Santé et de la Recherche Médicale (INSERM), U1292, MAB2 Team, Laboratoire de BioSanté, Bât C3, Pièce 304B.17 rue des Martyrs, 38054, Grenoble, France
- Commissariat à l'Energie Atomique (CEA), DSV-IRIG, Grenoble, France
- Université Grenoble Alpes (UGA), Grenoble, France
| | - Mohamed Benharouga
- Institut National de La Santé et de la Recherche Médicale (INSERM), U1292, MAB2 Team, Laboratoire de BioSanté, Bât C3, Pièce 304B.17 rue des Martyrs, 38054, Grenoble, France
- Commissariat à l'Energie Atomique (CEA), DSV-IRIG, Grenoble, France
- Université Grenoble Alpes (UGA), Grenoble, France
| | - Antoine Giraud
- INSERM U1059 SAINBIOSE, Université Jean Monnet, Saint-Étienne, France
- Neonatal Intensive Care Unit, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Étienne, France
| | - Nadia Alfaidy
- Institut National de La Santé et de la Recherche Médicale (INSERM), U1292, MAB2 Team, Laboratoire de BioSanté, Bât C3, Pièce 304B.17 rue des Martyrs, 38054, Grenoble, France.
- Commissariat à l'Energie Atomique (CEA), DSV-IRIG, Grenoble, France.
- Université Grenoble Alpes (UGA), Grenoble, France.
| |
Collapse
|
16
|
Amodeo G, Franchi S, Galimberti G, Riboldi B, Sacerdote P. The Prokineticin System in Inflammatory Bowel Diseases: A Clinical and Preclinical Overview. Biomedicines 2023; 11:2985. [PMID: 38001985 PMCID: PMC10669895 DOI: 10.3390/biomedicines11112985] [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: 09/26/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Inflammatory bowel disease (IBD) includes Crohn's disease (CD) and ulcerative colitis (UC), which are characterized by chronic inflammation of the gastrointestinal (GI) tract. IBDs clinical manifestations are heterogeneous and characterized by a chronic relapsing-remitting course. Typical gastrointestinal signs and symptoms include diarrhea, GI bleeding, weight loss, and abdominal pain. Moreover, the presence of pain often manifests in the remitting disease phase. As a result, patients report a further reduction in life quality. Despite the scientific advances implemented in the last two decades and the therapies aimed at inducing or maintaining IBDs in a remissive condition, to date, their pathophysiology still remains unknown. In this scenario, the importance of identifying a common and effective therapeutic target for both digestive symptoms and pain remains a priority. Recent clinical and preclinical studies have reported the prokineticin system (PKS) as an emerging therapeutic target for IBDs. PKS alterations are likely to play a role in IBDs at multiple levels, such as in intestinal motility, local inflammation, ulceration processes, localized abdominal and visceral pain, as well as central nervous system sensitization, leading to the development of chronic and widespread pain. This narrative review summarized the evidence about the involvement of the PKS in IBD and discussed its potential as a druggable target.
Collapse
Affiliation(s)
- Giada Amodeo
- Dipartimento di Scienze Farmacologiche e Biomolecolari “Rodolfo Paoletti”, University of Milan, Via Vanvitelli 32, 20129 Milan, Italy; (S.F.); (G.G.); (B.R.); (P.S.)
| | | | | | | | | |
Collapse
|
17
|
Vincenzi M, Kremić A, Jouve A, Lattanzi R, Miele R, Benharouga M, Alfaidy N, Migrenne-Li S, Kanthasamy AG, Porcionatto M, Ferrara N, Tetko IV, Désaubry L, Nebigil CG. Therapeutic Potential of Targeting Prokineticin Receptors in Diseases. Pharmacol Rev 2023; 75:1167-1199. [PMID: 37684054 PMCID: PMC10595023 DOI: 10.1124/pharmrev.122.000801] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 09/10/2023] Open
Abstract
The prokineticins (PKs) were discovered approximately 20 years ago as small peptides inducing gut contractility. Today, they are established as angiogenic, anorectic, and proinflammatory cytokines, chemokines, hormones, and neuropeptides involved in variety of physiologic and pathophysiological pathways. Their altered expression or mutations implicated in several diseases make them a potential biomarker. Their G-protein coupled receptors, PKR1 and PKR2, have divergent roles that can be therapeutic target for treatment of cardiovascular, metabolic, and neural diseases as well as pain and cancer. This article reviews and summarizes our current knowledge of PK family functions from development of heart and brain to regulation of homeostasis in health and diseases. Finally, the review summarizes the established roles of the endogenous peptides, synthetic peptides and the selective ligands of PKR1 and PKR2, and nonpeptide orthostatic and allosteric modulator of the receptors in preclinical disease models. The present review emphasizes the ambiguous aspects and gaps in our knowledge of functions of PKR ligands and elucidates future perspectives for PK research. SIGNIFICANCE STATEMENT: This review provides an in-depth view of the prokineticin family and PK receptors that can be active without their endogenous ligand and exhibits "constitutive" activity in diseases. Their non- peptide ligands display promising effects in several preclinical disease models. PKs can be the diagnostic biomarker of several diseases. A thorough understanding of the role of prokineticin family and their receptor types in health and diseases is critical to develop novel therapeutic strategies with safety concerns.
Collapse
Affiliation(s)
- Martina Vincenzi
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Amin Kremić
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Appoline Jouve
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Roberta Lattanzi
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Rossella Miele
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Mohamed Benharouga
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Nadia Alfaidy
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Stephanie Migrenne-Li
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Anumantha G Kanthasamy
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Marimelia Porcionatto
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Napoleone Ferrara
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Igor V Tetko
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Laurent Désaubry
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| | - Canan G Nebigil
- Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Center of Research in Biomedicine of Strasbourg, Strasbourg, France (M.V., A.K., A.J., L.D., C.G.N.); Department of Physiology and Pharmacology (M.V., R.L.), and Department of Biochemical Sciences "Alessandro Rossi Fanelli" (R.M.), Sapienza University of Rome, Rome, Italy; University Grenoble Alpes, INSERM, CEA, Grenoble, France (M.B., N.A.); Unité de Biologie Fonctionnelle et Adaptative, Université Paris Cité, CNRS, Paris, France (S.M.); Department of Physiology and Pharamacology, Center for Neurologic Disease Research, University of Georgia, Athens, Georgia (A.G.K.); Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil (M.A.P.); Moores Cancer Center, University of California, San Diego, La Jolla, California (N.F.); and Institute of Structural Biology, Helmholtz Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany (I.V.T.); and BIGCHEM GmbH, Valerystr. 49, Unterschleissheim, Germany (I.V.T.)
| |
Collapse
|
18
|
Pérez-Gutiérrez L, Ferrara N. Biology and therapeutic targeting of vascular endothelial growth factor A. Nat Rev Mol Cell Biol 2023; 24:816-834. [PMID: 37491579 DOI: 10.1038/s41580-023-00631-w] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2023] [Indexed: 07/27/2023]
Abstract
The formation of new blood vessels, called angiogenesis, is an essential pathophysiological process in which several families of regulators have been implicated. Among these, vascular endothelial growth factor A (VEGFA; also known as VEGF) and its two tyrosine kinase receptors, VEGFR1 and VEGFR2, represent a key signalling pathway mediating physiological angiogenesis and are also major therapeutic targets. VEGFA is a member of the gene family that includes VEGFB, VEGFC, VEGFD and placental growth factor (PLGF). Three decades after its initial isolation and cloning, VEGFA is arguably the most extensively investigated signalling system in angiogenesis. Although many mediators of angiogenesis have been identified, including members of the FGF family, angiopoietins, TGFβ and sphingosine 1-phosphate, all current FDA-approved anti-angiogenic drugs target the VEGF pathway. Anti-VEGF agents are widely used in oncology and, in combination with chemotherapy or immunotherapy, are now the standard of care in multiple malignancies. Anti-VEGF drugs have also revolutionized the treatment of neovascular eye disorders such as age-related macular degeneration and ischaemic retinal disorders. In this Review, we emphasize the molecular, structural and cellular basis of VEGFA action as well as recent findings illustrating unexpected interactions with other pathways and provocative reports on the role of VEGFA in regenerative medicine. We also discuss clinical and translational aspects of VEGFA. Given the crucial role that VEGFA plays in regulating angiogenesis in health and disease, this molecule is largely the focus of this Review.
Collapse
Affiliation(s)
- Lorena Pérez-Gutiérrez
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Napoleone Ferrara
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
19
|
Younes H, Kyritsi I, Mahrougui Z, Benharouga M, Alfaidy N, Marquette C. Effects of Prokineticins on Cerebral Cell Function and Blood-Brain Barrier Permeability. Int J Mol Sci 2023; 24:15428. [PMID: 37895111 PMCID: PMC10607385 DOI: 10.3390/ijms242015428] [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: 09/06/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Prokineticins are a family of small proteins with diverse roles in various tissues, including the brain. However, their specific effects on different cerebral cell types and blood-brain barrier (BBB) function remain unclear. The aim of this study was to investigate the effects of PROK1 and PROK2 on murine cerebral cell lines, bEnd.3, C8.D30, and N2a, corresponding to microvascular endothelial cells, astrocytes and neurons, respectively, and on an established BBB co-culture model. Western blot analysis showed that prokineticin receptors (PROKR1 and PROKR2) were differentially expressed in the considered cell lines. The effect of PROK1 and PROK2 on cell proliferation and migration were assessed using time-lapse microscopy. PROK1 decreased neural cells' proliferation, while it had no effect on the proliferation of endothelial cells and astrocytes. In contrast, PROK2 reduced the proliferation of all cell lines tested. Both PROK1 and PROK2 increased the migration of all cell lines. Blocking PROKRs with the PROKR1 antagonist (PC7) and the PROKR2 antagonist (PKR-A) inhibited astrocyte PROK2-mediated migration. Using the insert co-culture model of BBB, we demonstrated that PROKs increased BBB permeability, which could be prevented by PROKRs' antagonists.
Collapse
Affiliation(s)
- Hadi Younes
- University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France; (H.Y.); (I.K.); (Z.M.); (M.B.); (N.A.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Laboratory of Biology & Biotechnology for Health, Interdisciplinary Research Institute of Grenoble, 38000 Grenoble, France
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France
| | - Ioanna Kyritsi
- University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France; (H.Y.); (I.K.); (Z.M.); (M.B.); (N.A.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Laboratory of Biology & Biotechnology for Health, Interdisciplinary Research Institute of Grenoble, 38000 Grenoble, France
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France
| | - Zineb Mahrougui
- University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France; (H.Y.); (I.K.); (Z.M.); (M.B.); (N.A.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Laboratory of Biology & Biotechnology for Health, Interdisciplinary Research Institute of Grenoble, 38000 Grenoble, France
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France
| | - Mohamed Benharouga
- University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France; (H.Y.); (I.K.); (Z.M.); (M.B.); (N.A.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Laboratory of Biology & Biotechnology for Health, Interdisciplinary Research Institute of Grenoble, 38000 Grenoble, France
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France
| | - Nadia Alfaidy
- University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France; (H.Y.); (I.K.); (Z.M.); (M.B.); (N.A.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Laboratory of Biology & Biotechnology for Health, Interdisciplinary Research Institute of Grenoble, 38000 Grenoble, France
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France
| | - Christel Marquette
- University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France; (H.Y.); (I.K.); (Z.M.); (M.B.); (N.A.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Laboratory of Biology & Biotechnology for Health, Interdisciplinary Research Institute of Grenoble, 38000 Grenoble, France
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France
| |
Collapse
|
20
|
Onodera K, Tsuno Y, Hiraoka Y, Tanaka K, Maejima T, Mieda M. In vivo recording of the circadian calcium rhythm in Prokineticin 2 neurons of the suprachiasmatic nucleus. Sci Rep 2023; 13:16974. [PMID: 37813987 PMCID: PMC10562406 DOI: 10.1038/s41598-023-44282-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/05/2023] [Indexed: 10/11/2023] Open
Abstract
Prokineticin 2 (Prok2) is a small protein expressed in a subpopulation of neurons in the suprachiasmatic nucleus (SCN), the primary circadian pacemaker in mammals. Prok2 has been implicated as a candidate output molecule from the SCN to control multiple circadian rhythms. Genetic manipulation specific to Prok2-producing neurons would be a powerful approach to understanding their function. Here, we report the generation of Prok2-tTA knock-in mice expressing the tetracycline transactivator (tTA) specifically in Prok2 neurons and an application of these mice to in vivo recording of Ca2+ rhythms in these neurons. First, the specific and efficient expression of tTA in Prok2 neurons was verified by crossing the mice with EGFP reporter mice. Prok2-tTA mice were then used to express a fluorescent Ca2+ sensor protein to record the circadian Ca2+ rhythm in SCN Prok2 neurons in vivo. Ca2+ in these cells showed clear circadian rhythms in both light-dark and constant dark conditions, with their peaks around midday. Notably, the hours of high Ca2+ nearly coincided with the rest period of the behavioral rhythm. These observations fit well with the predicted function of Prok2 neurons as a candidate output pathway of the SCN by suppressing locomotor activity during both daytime and subjective daytime.
Collapse
Affiliation(s)
- Kaito Onodera
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Yusuke Tsuno
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Yuichi Hiraoka
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kohichi Tanaka
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takashi Maejima
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Michihiro Mieda
- Department of Integrative Neurophysiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan.
| |
Collapse
|
21
|
Rezaee A, Ahmadpour S, Jafari A, Aghili S, Zadeh SST, Rajabi A, Raisi A, Hamblin MR, Mahjoubin-Tehran M, Derakhshan M. MicroRNAs, long non-coding RNAs, and circular RNAs and gynecological cancers: focus on metastasis. Front Oncol 2023; 13:1215194. [PMID: 37854681 PMCID: PMC10580988 DOI: 10.3389/fonc.2023.1215194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/28/2023] [Indexed: 10/20/2023] Open
Abstract
Gynecologic cancer is a significant cause of death in women worldwide, with cervical cancer, ovarian cancer, and endometrial cancer being among the most well-known types. The initiation and progression of gynecologic cancers involve a variety of biological functions, including angiogenesis and metastasis-given that death mostly occurs from metastatic tumors that have invaded the surrounding tissues. Therefore, understanding the molecular pathways underlying gynecologic cancer metastasis is critical for enhancing patient survival and outcomes. Recent research has revealed the contribution of numerous non-coding RNAs (ncRNAs) to metastasis and invasion of gynecologic cancer by affecting specific cellular pathways. This review focuses on three types of gynecologic cancer (ovarian, endometrial, and cervical) and three kinds of ncRNAs (long non-coding RNAs, microRNAs, and circular RNAs). We summarize the detailed role of non-coding RNAs in the different pathways and molecular interactions involved in the invasion and metastasis of these cancers.
Collapse
Affiliation(s)
- Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Ahmadpour
- Biotechnology Department, Faculty of Chemistry, University of Kashan, Kashan, Iran
| | - Ameneh Jafari
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sarehnaz Aghili
- Department of Gynecology and Obstetrics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Ali Rajabi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Arash Raisi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Maryam Mahjoubin-Tehran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Derakhshan
- Shahid Beheshti Fertility Clinic, Department of Gynecology and Obsteterics, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
22
|
Cao Y, Langer R, Ferrara N. Targeting angiogenesis in oncology, ophthalmology and beyond. Nat Rev Drug Discov 2023; 22:476-495. [PMID: 37041221 DOI: 10.1038/s41573-023-00671-z] [Citation(s) in RCA: 122] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2023] [Indexed: 04/13/2023]
Abstract
Angiogenesis is an essential process in normal development and in adult physiology, but can be disrupted in numerous diseases. The concept of targeting angiogenesis for treating diseases was proposed more than 50 years ago, and the first two drugs targeting vascular endothelial growth factor (VEGF), bevacizumab and pegaptanib, were approved in 2004 for the treatment of cancer and neovascular ophthalmic diseases, respectively. Since then, nearly 20 years of clinical experience with anti-angiogenic drugs (AADs) have demonstrated the importance of this therapeutic modality for these disorders. However, there is a need to improve clinical outcomes by enhancing therapeutic efficacy, overcoming drug resistance, defining surrogate markers, combining with other drugs and developing the next generation of therapeutics. In this Review, we examine emerging new targets, the development of new drugs and challenging issues such as the mode of action of AADs and elucidating mechanisms underlying clinical benefits; we also discuss possible future directions of the field.
Collapse
Affiliation(s)
- Yihai Cao
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institute, Stockholm, Sweden.
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Napoleone Ferrara
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
23
|
Parab S, Setten E, Astanina E, Bussolino F, Doronzo G. The tissue-specific transcriptional landscape underlines the involvement of endothelial cells in health and disease. Pharmacol Ther 2023; 246:108418. [PMID: 37088448 DOI: 10.1016/j.pharmthera.2023.108418] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 03/23/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
Endothelial cells (ECs) that line vascular and lymphatic vessels are being increasingly recognized as important to organ function in health and disease. ECs participate not only in the trafficking of gases, metabolites, and cells between the bloodstream and tissues but also in the angiocrine-based induction of heterogeneous parenchymal cells, which are unique to their specific tissue functions. The molecular mechanisms regulating EC heterogeneity between and within different tissues are modeled during embryogenesis and become fully established in adults. Any changes in adult tissue homeostasis induced by aging, stress conditions, and various noxae may reshape EC heterogeneity and induce specific transcriptional features that condition a functional phenotype. Heterogeneity is sustained via specific genetic programs organized through the combinatory effects of a discrete number of transcription factors (TFs) that, at the single tissue-level, constitute dynamic networks that are post-transcriptionally and epigenetically regulated. This review is focused on outlining the TF-based networks involved in EC specialization and physiological and pathological stressors thought to modify their architecture.
Collapse
Affiliation(s)
- Sushant Parab
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Elisa Setten
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Elena Astanina
- Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy.
| | - Gabriella Doronzo
- Department of Oncology, University of Torino, IT, Italy; Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Torino, IT, Italy
| |
Collapse
|
24
|
Leu T, Denda J, Wrobeln A, Fandrey J. Hypoxia-Inducible Factor-2alpha Affects the MEK/ERK Signaling Pathway via Primary Cilia in Connection with the Intraflagellar Transport Protein 88 Homolog. Mol Cell Biol 2023; 43:174-183. [PMID: 37074220 PMCID: PMC10153011 DOI: 10.1080/10985549.2023.2198931] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/25/2023] [Indexed: 04/20/2023] Open
Abstract
The ability of cells to communicate with their surrounding is a prerequisite for essential processes such as proliferation, apoptosis, migration, and differentiation. To this purpose, primary cilia serve as antennae-like structures on the surface of most mammalian cell types. Cilia allow signaling via hedgehog, Wnt or TGF-beta pathways. Their length, in part controlled by the activity of intraflagellar transport (IFT), is a parameter for adequate function of primary cilia. Here we show, in murine neuronal cells, that intraflagellar transport protein 88 homolog (IFT88) directly interacts with the hypoxia-inducible factor-2α (HIF-2α), hitherto known as an oxygen-regulated transcription factor. Furthermore, HIF-2α accumulates in the ciliary axoneme and promotes ciliary elongation under hypoxia. Loss of HIF-2α affected ciliary signaling in neuronal cells by decreasing transcription of Mek1/2 and Erk1/2. Targets of the MEK/ERK signaling pathway, such as Fos and Jun, were significantly decreased. Our results suggest that HIF-2α influences ciliary signaling by interacting with IFT88 under hypoxic conditions. This implies an unexpected and far more extensive function of HIF-2α than described before.
Collapse
Affiliation(s)
- Tristan Leu
- Institute of Physiology, University Duisburg-Essen, Essen, Germany
| | - Jannik Denda
- Institute of Physiology, University Duisburg-Essen, Essen, Germany
| | - Anna Wrobeln
- Institute of Physiology, University Duisburg-Essen, Essen, Germany
| | - Joachim Fandrey
- Institute of Physiology, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
25
|
Baryla M, Goryszewska-Szczurek E, Kaczynski P, Balboni G, Waclawik A. Prokineticin 1 is a novel factor regulating porcine corpus luteum function. Sci Rep 2023; 13:5085. [PMID: 36991037 PMCID: PMC10060428 DOI: 10.1038/s41598-023-32132-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
Prokineticin 1 (PROK1) is a pleiotropic factor secreted by endocrine glands; however, its role has not been studied in the corpus luteum (CL) during pregnancy in any species. The present study aimed to investigate the contribution of PROK1 in regulating processes related to porcine CL function and regression: steroidogenesis, luteal cell apoptosis and viability, and angiogenesis. The luteal expression of PROK1 was greater on Days 12 and 14 of pregnancy compared to Day 9. PROK1 protein expression during pregnancy increased gradually and peaked on Day 14, when it was also significantly higher than that on Day 14 of the estrous cycle. Prokineticin receptor 1 (PROKR1) mRNA abundance increased on Days 12 and 14 of pregnancy, whereas PROKR2 elevated on Day 14 of the estrous cycle. PROK1, acting via PROKR1, stimulated the expression of genes involved in progesterone synthesis, as well as progesterone secretion by luteal tissue. PROK1-PROKR1 signaling reduced apoptosis and increased the viability of luteal cells. PROK1 acting through PROKR1 stimulated angiogenesis by increasing capillary-like structure formation by luteal endothelial cells and elevating angiogenin gene expression and VEGFA secretion by luteal tissue. Our results indicate that PROK1 regulates processes vital for maintaining luteal function during early pregnancy and the mid-luteal phase.
Collapse
Affiliation(s)
- Monika Baryla
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Ewelina Goryszewska-Szczurek
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Piotr Kaczynski
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Gianfranco Balboni
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria, 09042, Monserrato, Cagliari, Italy
| | - Agnieszka Waclawik
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| |
Collapse
|
26
|
Lattanzi R, Miele R. Non-Peptide Agonists and Antagonists of the Prokineticin Receptors. Curr Issues Mol Biol 2022; 44:6323-6332. [PMID: 36547092 PMCID: PMC9776816 DOI: 10.3390/cimb44120431] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
The prokineticin family comprises a group of secreted peptides that can be classified as chemokines based on their structural features and chemotactic and immunomodulatory functions. Prokineticins (PKs) bind with high affinity to two G protein-coupled receptors (GPCRs). Prokineticin receptor 1 (PKR1) and prokineticin receptor 2 (PKR2) are involved in a variety of physiological functions such as angiogenesis and neurogenesis, hematopoiesis, the control of hypothalamic hormone secretion, the regulation of circadian rhythm and the modulation of complex behaviors such as feeding and drinking. Dysregulation of the system leads to an inflammatory process that is the substrate for many pathological conditions such as cancer, pain, neuroinflammation and neurodegenerative diseases such as Alzheimer's and Parkinson's disease. The use of PKR's antagonists reduces PK2/PKRs upregulation triggered by various inflammatory processes, suggesting that a pharmacological blockade of PKRs may be a successful strategy to treat inflammatory/neuroinflammatory diseases, at least in rodents. Under certain circumstances, the PK system exhibits protective/neuroprotective effects, so PKR agonists have also been developed to modulate the prokineticin system.
Collapse
Affiliation(s)
- Roberta Lattanzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Rossella Miele
- Department of Biochemical Sciences “A. Rossi Fanelli”, CNR-Institute of Molecular Biology and Pathology, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| |
Collapse
|
27
|
Targeting hypoxia-related metabolism molecules: How to improve tumour immune and clinical treatment? Biomed Pharmacother 2022; 156:113917. [DOI: 10.1016/j.biopha.2022.113917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/20/2022] Open
|
28
|
Espes D, Magnusson L, Caballero-Corbalan J, Schwarcz E, Casas R, Carlsson PO. Pregnancy induces pancreatic insulin secretion in women with long-standing type 1 diabetes. BMJ Open Diabetes Res Care 2022; 10:10/6/e002948. [PMID: 36351678 PMCID: PMC9644305 DOI: 10.1136/bmjdrc-2022-002948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 10/01/2022] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Pregnancy entails both pancreatic adaptations with increasing β-cell mass and immunological alterations in healthy women. In this study, we have examined the effects of pregnancy on β-cell function and immunological processes in long-standing type 1 diabetes (L-T1D). RESEARCH DESIGN AND METHODS Fasting and stimulated C-peptide were measured after an oral glucose tolerance test in pregnant women with L-T1D (n=17) during the first trimester, third trimester, and 5-8 weeks post partum. Two 92-plex Olink panels were used to measure proteins in plasma. Non-pregnant women with L-T1D (n=30) were included for comparison. RESULTS Fasting C-peptide was detected to a higher degree in women with L-T1D during gestation and after parturition (first trimester: 64.7%, third trimester: 76.5%, and post partum: 64.7% vs 26.7% in non-pregnant women). Also, total insulin secretion and peak C-peptide increased during pregnancy. The plasma protein levels in pregnant women with L-T1D was dynamic, but few analytes were functionally related. Specifically, peripheral levels of prolactin (PRL), prokineticin (PROK)-1, and glucagon (GCG) were elevated during gestation whereas levels of proteins related to leukocyte migration (CCL11), T cell activation (CD28), and antigen presentation (such as CD83) were reduced. CONCLUSIONS In summary, we have found that some C-peptide secretion, that is, an indirect measurement of endogenous insulin production, is regained in women with L-T1D during pregnancy, which might be attributed to elevated peripheral levels of PRL, PROK-1, or GCG.
Collapse
Affiliation(s)
- Daniel Espes
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Louise Magnusson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linkoping, Sweden
| | | | - Erik Schwarcz
- Department of Internal Medicine, Örebro University Hospital, Orebro, Sweden
| | - Rosaura Casas
- Department of Biomedical and Clinical Sciences, Linköping University, Linkoping, Sweden
| | - Per-Ola Carlsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
29
|
Prokineticins as a Prognostic Biomarker for Low-Grade Gliomas: A Study Based on The Cancer Genome Atlas Data. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2309339. [PMID: 35845958 PMCID: PMC9283042 DOI: 10.1155/2022/2309339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/23/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022]
Abstract
Lower-grade glioma (LGG) is a crucial pathological type of glioma. Prokineticins have not been reported in LGG. Prokineticins as a member of the multifunctional chemokine-like peptide family are divided into two ligands: PROK1 and PROK2. We evaluated the role of PROK1 and PROK2 in LGG using TCGA database. We downloaded the datasets of LGG from TCGA and evaluated the influence of prokineticins on LGG survival by survival module. Correlations between clinical information and prokineticins expression were analyzed using logistic regression. Univariable survival and multivariate Cox analysis was used to compare several clinical characteristics with survival. Correlation between prokineticins and cancer immune infiltrates was explored using CIBERSORT and correlation module of GEPIA. We analyzed genes of PROK1 and PROK2 affecting LGG, screened differentially expressed genes (DEGs), interacted protein-protein with DEGs through the STRING website, then imported the results into the Cytospace software, and calculated the hub genes. To analyze whether hub genes and prokineticins are related, the relationship between PROK1 and PROK2 and hub genes was assessed and shown by heat map. In addition, gene set enrichment analysis (GSEA) was performed using the TCGA dataset. The univariate analysis using logistic regression and PROK1 and PROK2 showed opposite expression differences between tumor and normal tissues (
). PRO1 and PROK2 expressions showed significant differences in tumor grade, age, Iiscitrate DeHydrogenase (IDH) status, histological type, and 1P/19q codeletion. Multivariate analysis revealed that the up-regulated PROK1 and PROK2 expression is an independent prognostic factor for bad prognosis. Specifically, prokineticin expression level has significant correlations with infiltrating levels of Th1 cells, NK CD 56bright cells, and Mast cells in LGG. We screened 21 DEGs and obtained 5 hub genes (HOXC10, HOXD13, SOX4, GATA4, HOXA9). GSEA-identified FCMR activation, creation of C4 and C2 activators, and CD22-mediated BCR regulation in gene ontology (GO) were differentially enriched in high PROK1 and PROK2 expression phenotype pathway, cytoplasmic ribosomal proteins, and ribosome and were differentially enriched in the low PROK1 and PROK2 expression phenotype pathway. Prokineticins are a prognostic biomarker and the correlation between hub genes and LGG requires further attention.
Collapse
|
30
|
Lattanzi R, Severini C, Miele R. Prokineticin 2 in cancer-related inflammation. Cancer Lett 2022; 546:215838. [DOI: 10.1016/j.canlet.2022.215838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 11/28/2022]
|
31
|
Puliani G, Sesti F, Anastasi E, Verrico M, Tarsitano MG, Feola T, Campolo F, Di Gioia CRT, Venneri MA, Angeloni A, Appetecchia M, Lenzi A, Isidori AM, Faggiano A, Giannetta E. Angiogenic factors as prognostic markers in neuroendocrine neoplasms. Endocrine 2022; 76:208-217. [PMID: 35088292 DOI: 10.1007/s12020-021-02942-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/07/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE Angiogenic markers in neuroendocrine neoplasms (NENs) have recently received increasing attention, but their clinical role remains unclear. The aim of this study was to evaluate the role of angiogenic markers in NEN aggressiveness and prognosis. METHODS We performed a prospective observational study including 46 consecutive patients with proven NENs of pulmonary (45.65%) and gastro-entero-pancreatic (GEP) (54.35%) origin and 29 healthy controls. Circulating pro-angiogenic factors were measured by ELISA assay. ANG2 tissue expression was evaluated in a subgroup of ten patients by immunohistochemistry. RESULTS The study demonstrated a significantly higher level of ANG2, ANG1, sTIE2, and PROK2 in patients affected by NENs compared to controls. In the NENs' group we measured that: (i) ANG2 levels were higher in poorly vs well-differentiated NENs: 4.85 (2.75-7.42) vs 3.16 (1.66-6.36) ng/ml, p = 0.046 and in tumor stage 3-4 compared to stage 1-2: 4.24 (2.66-8.72) vs 2.73 (1.53-5.70), p = 0.044; (ii) ANG2 and PROK2 were significantly higher in patents with progressive disease compared to stable disease: ANG2 = 6.26 (3.98-10.99) vs 2.73 (1.65-4.36) pg/ml, p = 0.001; PROK2 = 29.19 (28.42-32.25) vs 28.37 (28.14-28.91) pg/ml, p = 0.035. Immunohistochemistry confirmed ANG2 expression in tumor specimens. CONCLUSIONS We documented higher levels of angiogenic markers in NENs, with an association between ANG2 serum levels and NENs morphology and staging. In both GEP and lung NENs, ANG2 and PROK2 are higher in case of tumor progression, suggesting a potential role as prognostic markers in NENs patients.
Collapse
Affiliation(s)
- Giulia Puliani
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Franz Sesti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Emanuela Anastasi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Monica Verrico
- Medical Oncology Unit A, Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | | | - Tiziana Feola
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
- Neuroendocrinology, Neuromed Institute, IRCCS, Pozzilli, Italy
| | - Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonio Angeloni
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Marialuisa Appetecchia
- Oncological Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Elisa Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
32
|
Van Baelen AC, Robin P, Kessler P, Maïga A, Gilles N, Servent D. Structural and Functional Diversity of Animal Toxins Interacting With GPCRs. Front Mol Biosci 2022; 9:811365. [PMID: 35198603 PMCID: PMC8859281 DOI: 10.3389/fmolb.2022.811365] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
Peptide toxins from venoms have undergone a long evolutionary process allowing host defense or prey capture and making them highly selective and potent for their target. This has resulted in the emergence of a large panel of toxins from a wide diversity of species, with varied structures and multiple associated biological functions. In this way, animal toxins constitute an inexhaustible reservoir of druggable molecules due to their interesting pharmacological properties. One of the most interesting classes of therapeutic targets is the G-protein coupled receptors (GPCRs). GPCRs represent the largest family of membrane receptors in mammals with approximately 800 different members. They are involved in almost all biological functions and are the target of almost 30% of drugs currently on the market. Given the interest of GPCRs in the therapeutic field, the study of toxins that can interact with and modulate their activity with the purpose of drug development is of particular importance. The present review focuses on toxins targeting GPCRs, including peptide-interacting receptors or aminergic receptors, with a particular focus on structural aspects and, when relevant, on potential medical applications. The toxins described here exhibit a great diversity in size, from 10 to 80 amino acids long, in disulfide bridges, from none to five, and belong to a large panel of structural scaffolds. Particular toxin structures developed here include inhibitory cystine knot (ICK), three-finger fold, and Kunitz-type toxins. We summarize current knowledge on the structural and functional diversity of toxins interacting with GPCRs, concerning first the agonist-mimicking toxins that act as endogenous agonists targeting the corresponding receptor, and second the toxins that differ structurally from natural agonists and which display agonist, antagonist, or allosteric properties.
Collapse
Affiliation(s)
- Anne-Cécile Van Baelen
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Philippe Robin
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Pascal Kessler
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Arhamatoulaye Maïga
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
- CHU Sainte Justine, Université de Montréal, Montreal, QC, Canada
| | - Nicolas Gilles
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Denis Servent
- CEA, Département Médicaments et Technologies pour La Santé (DMTS), SIMoS, Université Paris-Saclay, Gif-sur-Yvette, France
- *Correspondence: Denis Servent,
| |
Collapse
|
33
|
Marei I, Chidiac O, Thomas B, Pasquier J, Dargham S, Robay A, Vakayil M, Jameesh M, Triggle C, Rafii A, Jayyousi A, Al Suwaidi J, Abi Khalil C. Angiogenic content of microparticles in patients with diabetes and coronary artery disease predicts networks of endothelial dysfunction. Cardiovasc Diabetol 2022; 21:17. [PMID: 35109843 PMCID: PMC8812242 DOI: 10.1186/s12933-022-01449-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/20/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Elevated endothelial microparticles (EMPs) levels are surrogate markers of vascular dysfunction. We analyzed EMPs with apoptotic characteristics and assessed the angiogenic contents of microparticles in the blood of patients with type 2 diabetes (T2D) according to the presence of coronary artery disease (CAD). METHODS A total of 80 participants were recruited and equally classified as (1) healthy without T2D, (2) T2D without cardiovascular complications, (3) T2D and chronic coronary artery disease (CAD), and (4) T2D and acute coronary syndrome (ACS). MPs were isolated from the peripheral circulation, and EMPs were characterized using flow cytometry of CD42 and CD31. CD62E was used to determine EMPs' apoptotic/activation state. MPs content was extracted and profiled using an angiogenesis array. RESULTS Levels of CD42- CD31 + EMPs were significantly increased in T2D with ACS (257.5 ± 35.58) when compared to healthy subjects (105.7 ± 12.96, p < 0.01). There was no significant difference when comparing T2D with and without chronic CAD. The ratio of CD42-CD62 +/CD42-CD31 + EMPs was reduced in all T2D patients, with further reduction in ACS when compared to chronic CAD, reflecting a release by apoptotic endothelial cells. The angiogenic content of the full population of MPs was analyzed. It revealed a significant differential expression of 5 factors in patients with ACS and diabetes, including TGF-β1, PD-ECGF, platelet factor 4, serpin E1, and thrombospondin 1. Ingenuity Pathway Analysis revealed that those five differentially expressed molecules, mainly TGF-β1, inhibit key pathways involved in normal endothelial function. Further comparison of the three diabetes groups to healthy controls and diabetes without cardiovascular disease to diabetes with CAD identified networks that inhibit normal endothelial cell function. Interestingly, DDP-IV was the only differentially expressed protein between chronic CAD and ACS in patients with diabetes. CONCLUSION Our data showed that the release of apoptosis-induced EMPs is increased in diabetes, irrespective of CAD, ACS patients having the highest levels. The protein contents of MPs interact in networks that indicate vascular dysfunction.
Collapse
Affiliation(s)
- Isra Marei
- Department of Pharmacology, Weill Cornell Medicine-Qatar, Doha, Qatar
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Genetic Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Omar Chidiac
- Department of Genetic Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Binitha Thomas
- Department of Genetic Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Jennifer Pasquier
- Department of Genetic Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Soha Dargham
- Biostatistics Core, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Amal Robay
- Department of Genetic Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Muneera Vakayil
- Department of Genetic Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | | | | | - Arash Rafii
- Department of Genetic Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Amin Jayyousi
- Department of Endocrinology, Hamad Medical Corporation, Doha, Qatar
| | | | - Charbel Abi Khalil
- Department of Genetic Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar.
- Heart Hospital, Hamad Medical Corporation, Doha, Qatar.
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, USA.
| |
Collapse
|
34
|
Lian M, Shao S, Liu M, Shi Y, Zhang H, Chen D. Cell membrane-coated nanoparticles as peroxidase mimetics for cancer cell targeted detection and therapy. Talanta 2022; 238:123071. [PMID: 34808566 DOI: 10.1016/j.talanta.2021.123071] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/22/2021] [Accepted: 11/14/2021] [Indexed: 01/10/2023]
Abstract
The development of novel and efficient recognition molecules that can be easily modified by nanomaterials to achieve ultra-sensitive and specific cancer cell analysis is of great significance for its early diagnosis and timely prognosis. Herin, a new nanostructured hybrid based on cell membrane-coated Au cores- ultrathin Pt skins composite nanoparticles (Au@Pt@CM NPs) were developed for in vitro detection and treatment of cancer cells. In this strategy, the Au@Pt NPs acted as the signal transducer, and the cell membrane were used as the cancer-cell recognition tool. The synthesized Au@Pt@CM NPs could catalyze the oxidation of 3,3',5,5'-tetramethylbenzidine (TMB) in the presence of the hydrogen peroxide and were demonstrated to have excellent peroxidase-like activity. Coated with the source cancer cell membrane, the nanoparticles achieved highly specific self-recognition to the source cell. Therefore, the colorimetric method based on Au@Pt@CM NPs could detect the cancer cells in the linear range from 50 to 100000 cells/mL with a limit of detection of 5 cells/mL, which is much lower than other colorimetric detection methods. Afterwards, the nanoparticles as a mimetic enzyme were used for therapeutics of cancer cells through the ROS-mediated oxidative damage. Due to the change of the redox state in the cells by the Au@Pt@CM NPs, the hybrid can achieve the growth inhibitory effect and the selective killing effect on cancer cells. It can be expected that this novel hybrid membrane coating method will bring new insight into developing targeted nanomaterials for tumor treatment and detection.
Collapse
Affiliation(s)
- Meiling Lian
- Key Laboratory of Civil Aviation Thermal Hazards Prevention and Emergency Response, Civil Aviation University of China, Tianjin, 300300, China
| | - Shuaibin Shao
- Key Laboratory of Civil Aviation Thermal Hazards Prevention and Emergency Response, Civil Aviation University of China, Tianjin, 300300, China
| | - Meihan Liu
- Key Laboratory of Civil Aviation Thermal Hazards Prevention and Emergency Response, Civil Aviation University of China, Tianjin, 300300, China
| | - Yuqing Shi
- Key Laboratory of Civil Aviation Thermal Hazards Prevention and Emergency Response, Civil Aviation University of China, Tianjin, 300300, China
| | - Haijun Zhang
- Key Laboratory of Civil Aviation Thermal Hazards Prevention and Emergency Response, Civil Aviation University of China, Tianjin, 300300, China.
| | - Da Chen
- Key Laboratory of Civil Aviation Thermal Hazards Prevention and Emergency Response, Civil Aviation University of China, Tianjin, 300300, China.
| |
Collapse
|
35
|
Lattanzi R, Miele R. Prokineticin-Receptor Network: Mechanisms of Regulation. Life (Basel) 2022; 12:172. [PMID: 35207461 PMCID: PMC8877203 DOI: 10.3390/life12020172] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 12/17/2022] Open
Abstract
Prokineticins are a new class of chemokine-like peptides that bind their G protein-coupled receptors, PKR1 and PKR2, and promote chemotaxis and the production of pro-inflammatory cytokines following tissue injury or infection. This review summarizes the major cellular and biochemical mechanisms of prokineticins pathway regulation that, like other chemokines, include: genetic polymorphisms; mRNA splice modulation; expression regulation at transcriptional and post-transcriptional levels; prokineticins interactions with cell-surface glycosaminoglycans; PKRs degradation, localization, post-translational modifications and oligomerization; alternative signaling responses; binding to pharmacological inhibitors. Understanding these mechanisms, which together exert substantial biochemical control and greatly enhance the complexity of the prokineticin-receptor network, leads to novel opportunities for therapeutic intervention. In this way, besides targeting prokineticins or their receptors directly, it could be possible to indirectly influence their activity by modulating their expression and localization or blocking the downstream signaling pathways.
Collapse
Affiliation(s)
- Roberta Lattanzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Rossella Miele
- Department of Biochemical Sciences “A. Rossi Fanelli”, CNR-Institute of Molecular Biology and Pathology, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| |
Collapse
|
36
|
Li P, Li Q, Biswas N, Xin H, Diemer T, Liu L, Perez Gutierrez L, Paternostro G, Piermarocchi C, Domanskyi S, Wang RK, Ferrara N. LIF, a mitogen for choroidal endothelial cells, protects the choriocapillaris: implications for prevention of geographic atrophy. EMBO Mol Med 2022; 14:e14511. [PMID: 34779136 PMCID: PMC8749470 DOI: 10.15252/emmm.202114511] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/18/2022] Open
Abstract
In the course of our studies aiming to discover vascular bed-specific endothelial cell (EC) mitogens, we identified leukemia inhibitory factor (LIF) as a mitogen for bovine choroidal EC (BCE), although LIF has been mainly characterized as an EC growth inhibitor and an anti-angiogenic molecule. LIF stimulated growth of BCE while it inhibited, as previously reported, bovine aortic EC (BAE) growth. The JAK-STAT3 pathway mediated LIF actions in both BCE and BAE cells, but a caspase-independent proapoptotic signal mediated by cathepsins was triggered in BAE but not in BCE. LIF administration directly promoted activation of STAT3 and increased blood vessel density in mouse eyes. LIF also had protective effects on the choriocapillaris in a model of oxidative retinal injury. Analysis of available single-cell transcriptomic datasets shows strong expression of the specific LIF receptor in mouse and human choroidal EC. Our data suggest that LIF administration may be an innovative approach to prevent atrophy associated with AMD, through protection of the choriocapillaris.
Collapse
Affiliation(s)
- Pin Li
- Department of PathologyUniversity of California San DiegoLa JollaCAUSA
| | - Qin Li
- Department of OphthalmologyUniversity of California San DiegoLa JollaCAUSA
| | - Nilima Biswas
- Department of PathologyUniversity of California San DiegoLa JollaCAUSA
| | - Hong Xin
- Department of PathologyUniversity of California San DiegoLa JollaCAUSA
| | - Tanja Diemer
- Department of PathologyUniversity of California San DiegoLa JollaCAUSA
| | - Lixian Liu
- Department of PathologyUniversity of California San DiegoLa JollaCAUSA
| | | | | | - Carlo Piermarocchi
- Department of Physics and AstronomyMichigan State UniversityEast LansingMIUSA
| | - Sergii Domanskyi
- Department of Physics and AstronomyMichigan State UniversityEast LansingMIUSA
| | - Ruikang K Wang
- Department of BioengineeringUniversity of WashingtonSeattleWAUSA
| | - Napoleone Ferrara
- Department of PathologyUniversity of California San DiegoLa JollaCAUSA
- Department of OphthalmologyUniversity of California San DiegoLa JollaCAUSA
| |
Collapse
|
37
|
Tagai N, Goi T, Shimada M, Kurebayashi H. Plasma Prokineticin 1, a prognostic biomarker in colorectal cancer patients with curative resection: a retrospective cohort study. World J Surg Oncol 2021; 19:302. [PMID: 34657605 PMCID: PMC8522247 DOI: 10.1186/s12957-021-02421-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 10/07/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Prokineticin 1 (PROK1) was reported as an angiogenic factor, which is associated with tumor progression, cell invasion, and metastasis in colorectal cancer. Although the association between PROK1 expression in primary cancer lesion and patient prognosis was reported, it is unclear whether plasma PROK1 concentration may be a predictive factor in colorectal cancer patients. This study investigated the association between PROK1 concentration in plasma and prognosis in colorectal cancer patients. METHODS We measured preoperative PROK1 plasma levels using ELISA method, while PROK1 expression in primary cancer lesion was evaluated using immunohistochemistry (IHC). The association between plasma PROK1 levels and cancer-related survival rate (CRS) was evaluated. Additionally, we examined whether simultaneous PROK1 expression in both primary cancer lesions and plasma was correlated with CRS. The cancer-related survival rate was calculated using the Kaplan-Meier method, and survival estimates were compared using the log-rank test. RESULTS We have gathered eligible 130 CRC patients retrospectively. Out of 130 patients, 61 (46.9%) were positive on IHC in primary cancer, and 69 (53.1%) were negative, while 43 (33.1%) had high-value PROK1 in plasma. Out of these 43, 30 (25.4%) also had concomitant higher IHC expression in primary cancer. The plasma PROK1 levels tended to increase with advancing stages. The plasma PROK1-positive group had a lower 5-year CRS than the negative group (63.6% vs. 88.2%; P = 0.006). Additionally, simultaneous PROK1 expression was associated with a more significant decrease of 5-year CRS than both negative groups in all stages (76.2% vs. 92.5%; P = 0.003) and stage III (59.3% vs. 84.5%; P = 0.047). Multivariate analysis showed simultaneous PROK1 expression was independently associated with worse CRS (HR, 1.97; 95% CI 1.20‑3.24, P < 0.01). CONCLUSION PROK1 expression in preoperative plasma reflects poor prognosis in patients undergoing curative resection for colorectal cancer. The plasma PROK1 level may be a potential predictive marker, especially in stage III colorectal cancer patients.
Collapse
Affiliation(s)
- Noriyuki Tagai
- Department of Surgery 1, University of Fukui, 23-3 Matsuokashimoaizuki, Eiheiji-Cho, Yoshida-gun, Fukui, 910-1193, Japan.
| | - Takanori Goi
- Department of Surgery 1, University of Fukui, 23-3 Matsuokashimoaizuki, Eiheiji-Cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Michiaki Shimada
- Department of Surgery 1, University of Fukui, 23-3 Matsuokashimoaizuki, Eiheiji-Cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Hidetaka Kurebayashi
- Department of Surgery 1, University of Fukui, 23-3 Matsuokashimoaizuki, Eiheiji-Cho, Yoshida-gun, Fukui, 910-1193, Japan
| |
Collapse
|
38
|
Fiore M, Tarani L, Radicioni A, Spaziani M, Ferraguti G, Putotto C, Gabanella F, Maftei D, Lattanzi R, Minni A, Greco A, Tarani F, Petrella C. Serum Prokineticin-2 in Prepubertal and Adult Klinefelter Individuals. Can J Physiol Pharmacol 2021; 100:151-157. [PMID: 34614364 DOI: 10.1139/cjpp-2021-0457] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The prokineticin-2 (PROK2) is a small peptide belonging to the prokineticin family. In humans and rodents this chemokine is primarily involved in the control of central and peripheral reproductive processes. Klinefelter's syndrome (KS) is the first cause of male genetic infertility, due to an extra X chromosome, which may occur with a classical karyotype (47, XXY) or mosaic forms (46, XY/47, XXY). In affected subjects, pubertal maturation usually begins at an adequate chronological age, but when development is almost complete, they display a primary gonadal failure, with early spermatogenesis damage, and later onset of testosterone insufficiency. Thus, the main aim of the present study was to investigate the serum levels of PROK2 in prepubertal and adult KS patients, comparing them with healthy subjects. We showed for the first time the presence of PROK2 in the children serum but with significant changes in KS individuals. Indeed, compared to healthy subjects characterized by PROK2 serum elevation during the growth, KS individuals showed constant serum levels during the sexual maturation phase (higher during the prepubertal phase but lower during the adult age). In conclusion, these data indicate that in KS individuals PROK2 may be considered a biomarker for investigating the SK infertility process.
Collapse
Affiliation(s)
- Marco Fiore
- IBCN-CNR, Institute of Cell Biology and Neurobiology, Roma, Italy;
| | - Luigi Tarani
- "Sapienza" University of Rome, Department of Pediatrics, Rome, Italy;
| | - Antonio Radicioni
- Sapienza University of Rome, Department of Experimental Medicine, Rome, Italy;
| | - Matteo Spaziani
- Sapienza University of Rome, Department of Experimental Medicine, Rome, Italy;
| | - Giampiero Ferraguti
- Sapienza University of Rome, Department of Cellular Biotechnologies and Hematology, Rome, Italy;
| | - Carolina Putotto
- "Sapienza" University of Rome, Department of Pediatrics, rome, Italy;
| | - Francesca Gabanella
- IBBC-CNR), Rome, Italy.,Institute of Molecular Biology and Pathology (IBPM-CNR), Rome, Italy;
| | - Daniela Maftei
- Sapienza University of Rome, Department of Physiology and Pharmacology "Vittorio Erspamer", Rome, Italy;
| | - Roberta Lattanzi
- Sapienza University of Rome, Department of Physiology and Pharmacology "Vittorio Erspamer", Rome, Italy;
| | - Antonio Minni
- Sapienza University of Rome, Department of Sense Organs, Rome, Italy;
| | - Antonio Greco
- University of Rome La Sapienza, 9311, Rome, Lazio, Italy;
| | - Francesca Tarani
- "Sapienza" University of Rome, Department of Pediatrics, rome, Italy;
| | | |
Collapse
|
39
|
Naruse T, Goi T, Yamaguchi A. Prokineticin-1 induces normal lymphangiogenic activity and is involved in lymphangiogenesis and lymph node metastasis in colorectal cancer. Oncotarget 2021; 12:1388-1397. [PMID: 34262649 PMCID: PMC8274725 DOI: 10.18632/oncotarget.28016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/11/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Prokineticin family correlates with important roles in several biological processes, including homeostasis. We discovered novel functions of prokineticin1 (PROK1) in lymphangiogenesis and lymphnode metastasis in colorectal cancer. Materials and Methods: We examined changes in the number of lymphatic endothelial cells after PROK1 stimulation. PROK1 protein was stimulated with subcutaneously implanted in mice. Also a high-PROK1-expressing colorectal cancer cell line and anti-PROK1 antibody(Ab) were subcutaneously implanted in mice, and then examine lymphangiogenesis. PROK1 expression and the number of lymph vessels were examined in the primary lesion of 391 patients whose colorectal tumors had been resected. Results: When PROK1 was used as a stimulus, the number of lymphatic cells increased compared to unstimulated cells. And the number of lymph vessels in the skin of mice increased compared to mice implanted without PROK1. The number of lymph vessels in the primary tumor tissue increased when PROK1 was highly expressed compared to cases with non-detectable PROK1 expression. When PROK1 was expressed in human colorectal tumors, the rate of lymphnode metastasis was significantly higher than that in cases with non-detectable PROK1 expression. Conclusions: PROK1 is a lymphangiogenic factor involved in the formation of new lymph vessels and lymphnode metastasis in human colorectal cancer.
Collapse
Affiliation(s)
- Takayuki Naruse
- First Department of Surgery, University of Fukui, Fukui 9101193, Japan
| | - Takanori Goi
- First Department of Surgery, University of Fukui, Fukui 9101193, Japan
| | | |
Collapse
|
40
|
Rashid M, Zadeh LR, Baradaran B, Molavi O, Ghesmati Z, Sabzichi M, Ramezani F. Up-down regulation of HIF-1α in cancer progression. Gene 2021; 798:145796. [PMID: 34175393 DOI: 10.1016/j.gene.2021.145796] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/25/2021] [Accepted: 06/22/2021] [Indexed: 12/19/2022]
Abstract
Hypoxia induicible factor-1 alpha (HIF-1α) is a key transcription factor in cancer progression and target therapy in cancer. HIF-1α acts differently depending on presence or absence of Oxygen. In an oxygen-immersed environment, HIF-1α completely deactivated and destroyed by the ubiquitin proteasome pathway (UPP). In contrast, in the oxygen-free environment, it escapes destruction and enters to the nucleus of cells then upregulates many genes involved in cancer progression. Overexpressed HIF-1α and downstream genes support cancer progression through various mechanisms including angiogenesis, proliferation and survival of cells, metabolism reprogramming, invasion and metastasis, cancer stem cell maintenance, induction of genetic instability, and treatment resistance. HIF-1α can be provoked by signaling pathways unrelated to hypoxia during cancer progression. Therefore, cancer development and progression can be modulated by targeting HIF-1α and its downstream signaling molecules. In this regard, HIF-1α inhibitors which are categorized into the agents that regulate HIF-1α in gene, mRNA and protein levels used as an efficient way in cancer treatment. Also, HIF-1α expression can be negatively affected by the agents suppressing the activation of mTOR, PI3k/Akt and MAPK pathways.
Collapse
Affiliation(s)
- Mohsen Rashid
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Rostami Zadeh
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Ghesmati
- Department of Medical Biotechnology, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Sabzichi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Fatemeh Ramezani
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
41
|
Casella I, Ambrosio C. Prokineticin receptors interact unselectively with several G protein subtypes but bind selectively to β-arrestin 2. Cell Signal 2021; 83:110000. [PMID: 33811988 DOI: 10.1016/j.cellsig.2021.110000] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 11/19/2022]
Abstract
Prokineticin 1 (pk1) and prokineticin 2 (pk2) interact with two structurally related G-protein coupled receptors, prokineticin receptor 1 (PKR1) and prokineticin receptor 2 (PKR2). Cellular signalling studies show that the activated receptors can evoke Ca2+-mobilization, pertussis toxin-sensitive ERK phosphorylation, and intracellular cAMP accumulation, which suggests the partecipation of several G protein subtypes, such as Gq/11, Gi/o and Gs. However, direct interactions with these transduction proteins have not been studied yet. Here we measured by bioluminescence resonance energy transfer (BRET) the association of PKR1 and PKR2 with different heterotrimeric Gα proteins in response to pk1 and pk2 activation. Using host-cell lines carrying gene deletions of Gαq/11 or Gαs, and pertussis toxin treatment to abolish the receptor interactions with Gαi/o, we determined that both receptors could couple with comparable efficiency to Gq/11 and Gi/o, but far less efficiently to Gs or other pertussis toxin-insensitive G proteins. We also used BRET methodology to assess the association of prokineticin receptors with β-arrestin isoforms. Fluorescent versions of the isoforms were transfected both in HEK293 cells and in double KO β-arrestin 1/2 mouse fibroblasts, to study receptor interaction with the reconstituted individual β-arrestins without background expression of the endogenous genes. Both receptors formed stable BRET-emitting complexes with β-arrestin 2 but not with β-arrestin 1, indicating strong selectivity for the former. In all the studied transducer interactions and in both receptors, pk2 was more potent than pk1 in promoting receptor binding to transduction proteins.
Collapse
Affiliation(s)
- Ida Casella
- Istituto Superiore di Sanità, National Center for Drug Reserch and Evaluation, Viale Regina Elena, 299, 00161 Rome, Italy.
| | - Caterina Ambrosio
- Istituto Superiore di Sanità, National Center for Drug Reserch and Evaluation, Viale Regina Elena, 299, 00161 Rome, Italy
| |
Collapse
|
42
|
Reynaud D, Sergent F, Abi Nahed R, Traboulsi W, Collet C, Marquette C, Hoffmann P, Balboni G, Zhou QY, Murthi P, Benharouga M, Alfaidy N. Evidence-Based View of Safety and Effectiveness of Prokineticin Receptors Antagonists during Pregnancy. Biomedicines 2021; 9:309. [PMID: 33802771 PMCID: PMC8002561 DOI: 10.3390/biomedicines9030309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/04/2021] [Accepted: 03/13/2021] [Indexed: 12/13/2022] Open
Abstract
Endocrine gland derived vascular endothelial growth factor (EG-VEGF) is a canonical member of the prokineticin (PROKs) family. It acts via the two G-protein coupled receptors, namely PROKR1 and PROKR2. We have recently demonstrated that EG-VEGF is highly expressed in the human placenta; contributes to placental vascularization and growth and that its aberrant expression is associated with pregnancy pathologies including preeclampsia and fetal growth restriction. These findings strongly suggested that antagonization of its receptors may constitute a potential therapy for the pregnancy pathologies. Two specific antagonists of PROKR1 (PC7) and for PROKR2 (PKRA) were reported to reverse PROKs adverse effects in other systems. In the view of using these antagonists to treat pregnancy pathologies, a proof of concept study was designed to determine the biological significances of PC7 and PKRA in normal pregnancy outcome. PC7 and PKRA were tested independently or in combination in trophoblast cells and during early gestation in the gravid mouse. Both independent and combined treatments uncovered endogenous functions of EG-VEGF. The independent use of antagonists distinctively identified PROKR1 and PROKR2-mediated EG-VEGF signaling on trophoblast differentiation and invasion; thereby enhancing feto-placental growth and pregnancy outcome. Thus, our study provides evidence for the potential safe use of PC7 or PKRA to improve pregnancy outcome.
Collapse
Affiliation(s)
- Deborah Reynaud
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France; (D.R.); (F.S.); (R.A.N.); (C.C.); (C.M.); (P.H.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Frederic Sergent
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France; (D.R.); (F.S.); (R.A.N.); (C.C.); (C.M.); (P.H.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Roland Abi Nahed
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France; (D.R.); (F.S.); (R.A.N.); (C.C.); (C.M.); (P.H.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Wael Traboulsi
- Lombardi Comprehensive Cancer Center, Laboratory for Immuno-Oncology, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - Constance Collet
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France; (D.R.); (F.S.); (R.A.N.); (C.C.); (C.M.); (P.H.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Christel Marquette
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France; (D.R.); (F.S.); (R.A.N.); (C.C.); (C.M.); (P.H.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Pascale Hoffmann
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France; (D.R.); (F.S.); (R.A.N.); (C.C.); (C.M.); (P.H.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Gianfranco Balboni
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy;
| | - Qun-Yong Zhou
- Department of Pharmacology, University of California, Irvine, CA 92697, USA;
| | - Padma Murthi
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia;
- Department of Obstetrics and Gynecology, the University of Melbourne, Parkville, VIC 3010, Australia
| | - Mohamed Benharouga
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France; (D.R.); (F.S.); (R.A.N.); (C.C.); (C.M.); (P.H.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Nadia Alfaidy
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38000 Grenoble, France; (D.R.); (F.S.); (R.A.N.); (C.C.); (C.M.); (P.H.)
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| |
Collapse
|
43
|
Stucker S, De Angelis J, Kusumbe AP. Heterogeneity and Dynamics of Vasculature in the Endocrine System During Aging and Disease. Front Physiol 2021; 12:624928. [PMID: 33767633 PMCID: PMC7987104 DOI: 10.3389/fphys.2021.624928] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
The endocrine system consists of several highly vascularized glands that produce and secrete hormones to maintain body homeostasis and regulate a range of bodily functions and processes, including growth, metabolism and development. The dense and highly vascularized capillary network functions as the main transport system for hormones and regulatory factors to enable efficient endocrine function. The specialized capillary types provide the microenvironments to support stem and progenitor cells, by regulating their survival, maintenance and differentiation. Moreover, the vasculature interacts with endocrine cells supporting their endocrine function. However, the structure and niche function of vasculature in endocrine tissues remain poorly understood. Aging and endocrine disorders are associated with vascular perturbations. Understanding the cellular and molecular cues driving the disease, and age-related vascular perturbations hold potential to manage or even treat endocrine disorders and comorbidities associated with aging. This review aims to describe the structure and niche functions of the vasculature in various endocrine glands and define the vascular changes in aging and endocrine disorders.
Collapse
Affiliation(s)
| | | | - Anjali P. Kusumbe
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
| |
Collapse
|
44
|
Angiogenesis in the Normal Adrenal Fetal Cortex and Adrenocortical Tumors. Cancers (Basel) 2021; 13:cancers13051030. [PMID: 33804534 PMCID: PMC7957756 DOI: 10.3390/cancers13051030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 01/10/2023] Open
Abstract
Angiogenesis plays an important role in several physiological and pathological processes. Pharmacological angiogenesis modulation has been robustly demonstrated to achieve clinical benefits in several cancers. Adrenocortical carcinomas (ACC) are rare tumors that often have a poor prognosis. In addition, therapeutic options for ACC are limited. Understanding the mechanisms that regulate adrenocortical angiogenesis along the embryonic development and in ACC could provide important clues on how these processes could be pharmacologically modulated for ACC treatment. In this report, we performed an integrative review on adrenal cortex angiogenesis regulation in physiological conditions and ACC. During embryonic development, adrenal angiogenesis is regulated by both VEGF and Ang-Tie signaling pathways. In ACC, early research efforts were focused on VEGF signaling and this pathway was identified as a good prognostic factor and thus a promising therapeutic target. However, every clinical trial so far conducted in ACC using VEGF pathway- targeting drugs, alone or in combination, yielded disappointing results. In contrast, although the Ang-Tie pathway has been pointed out as an important regulator of fetal adrenocortical angiogenesis, its role is yet to be explored in ACC. In the future, further research on the role and efficacy of modulating both Ang-Tie and VEGF pathways in ACC is needed.
Collapse
|
45
|
Satija S, Kaur H, Tambuwala MM, Sharma P, Vyas M, Khurana N, Sharma N, Bakshi HA, Charbe NB, Zacconi FC, Aljabali AA, Nammi S, Dureja H, Singh TG, Gupta G, Dhanjal DS, Dua K, Chellappan DK, Mehta M. Hypoxia-Inducible Factor (HIF): Fuel for Cancer Progression. Curr Mol Pharmacol 2021; 14:321-332. [PMID: 33494692 DOI: 10.2174/1874467214666210120154929] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/24/2020] [Accepted: 10/09/2020] [Indexed: 11/22/2022]
Abstract
Hypoxia is an integral part of the tumor microenvironment, caused primarily due to rapidly multiplying tumor cells and a lack of proper blood supply. Among the major hypoxic pathways, HIF-1 transcription factor activation is one of the widely investigated pathways in the hypoxic tumor microenvironment (TME). HIF-1 is known to activate several adaptive reactions in response to oxygen deficiency in tumor cells. HIF-1 has two subunits, HIF-1β (constitutive) and HIF-1α (inducible). The HIF-1α expression is largely regulated via various cytokines (through PI3K-ACT-mTOR signals), which involves the cascading of several growth factors and oncogenic cascades. These events lead to the loss of cellular tumor suppressant activity through changes in the level of oxygen via oxygen-dependent and oxygen-independent pathways. The significant and crucial role of HIF in cancer progression and its underlying mechanisms have gained much attention lately among the translational researchers in the fields of cancer and biological sciences, which have enabled them to correlate these mechanisms with various other disease modalities. In the present review, we have summarized the key findings related to the role of HIF in the progression of tumors.
Collapse
Affiliation(s)
- Saurabh Satija
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Harpreet Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Prabal Sharma
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Manish Vyas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Navneet Khurana
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Neha Sharma
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Hamid A Bakshi
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, United Kingdom
| | - Nitin B Charbe
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Católica de Chile, Av. Vicuna McKenna 4860, 7820436 Macul, Santiago, Chile
| | - Flavia C Zacconi
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Católica de Chile, Av. Vicuna McKenna 4860, 7820436 Macul, Santiago, Chile
| | - Alaa A Aljabali
- Yarmouk University - Faculty of Pharmacy, Department of Pharmaceutical Sciences, Irbid, Jordan
| | - Srinivas Nammi
- School of Science and Health, Western Sydney University, Penrith NSW 2751, Australia
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Thakur G Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Gaurav Gupta
- School of Pharmaceutical Sciences, Suresh Gyan Vihar University, Jaipur, India
| | - Daljeet S Dhanjal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Dinesh K Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Meenu Mehta
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| |
Collapse
|
46
|
Chen C, Ge X, Zhao Y, Wang D, Ling L, Zheng S, Ding K, Wang J, Sun L. Molecular Alterations in Metastatic Ovarian Cancer From Gastrointestinal Cancer. Front Oncol 2020; 10:605349. [PMID: 33363035 PMCID: PMC7758447 DOI: 10.3389/fonc.2020.605349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/03/2020] [Indexed: 12/24/2022] Open
Abstract
Reports indicate that most metastatic ovarian cancer (MOC) originates from gastrointestinal cancer (GIC). Notably, GICs metastasize to the ovary frequently via 3 main routes including hematogenous spread, lymphogenous spread, and transcoelomic spread. Nonetheless, the mechanism of the progression remains unknown, and only a handful of literature exists on the molecular alteration implicated in MOC from GIC. This work collected existing evidence and literature on the vital molecules of the metastatic pathway and systematically analyzed them geared toward exploring the mechanism of the metastatic pathway of MOC. Further, this review described dominating molecular alteration in the metastatic process from cancer cells detaching away from lesions to arrive at the ovary, including factors for regulating signaling pathways in epithelial-interstitial transformation, invading, and surviving in the circulatory system or abdominal cavity. We interrogated the basis of the ovary as a distant metastatic site. This article provides new insights into the metastatic pathway and generates novel therapeutic targets for effective treatment and satisfactory outcomes in GIC patients.
Collapse
Affiliation(s)
- Chao Chen
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxu Ge
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yamei Zhao
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Da Wang
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Limian Ling
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu Zheng
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Wang
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lifeng Sun
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
47
|
Inhibition of protein glycosylation is a novel pro-angiogenic strategy that acts via activation of stress pathways. Nat Commun 2020; 11:6330. [PMID: 33303737 PMCID: PMC7730427 DOI: 10.1038/s41467-020-20108-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/11/2020] [Indexed: 01/05/2023] Open
Abstract
Endothelial cell (EC) metabolism is thought to be one of the driving forces for angiogenesis. Here we report the identification of the hexosamine D-mannosamine (ManN) as an EC mitogen and survival factor for bovine and human microvascular EC, with an additivity with VEGF. ManN inhibits glycosylation in ECs and induces significant changes in N-glycan and O-glycan profiles. We further demonstrate that ManN and two N-glycosylation inhibitors stimulate EC proliferation via both JNK activation and the unfolded protein response caused by ER stress. ManN results in enhanced angiogenesis in a mouse skin injury model. ManN also promotes angiogenesis in a mouse hindlimb ischemia model, with accelerated limb blood flow recovery compared to controls. In addition, intraocular injection of ManN induces retinal neovascularization. Therefore, activation of stress pathways following inhibition of protein glycosylation can promote EC proliferation and angiogenesis and may represent a therapeutic strategy for treatment of ischemic disorders. Therapeutic angiogenesis has the potential of inducing and maintaining new blood vessels and thus improving outcomes in patients with ischemic disorders. Mannosamine functions as an endothelial cell mitogen/survival factor through activation of stress pathways and might be useful to protect and regenerate the vascular endothelium in a variety of disorders.
Collapse
|
48
|
Alfaidy N, Brouillet S, Rajaraman G, Kalionis B, Hoffmann P, Barjat T, Benharouga M, Murthi P. The Emerging Role of the Prokineticins and Homeobox Genes in the Vascularization of the Placenta: Physiological and Pathological Aspects. Front Physiol 2020; 11:591850. [PMID: 33281622 PMCID: PMC7689260 DOI: 10.3389/fphys.2020.591850] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/13/2020] [Indexed: 01/25/2023] Open
Abstract
Vasculogenesis and angiogenesis are key processes of placental development, which occur throughout pregnancy. Placental vasculogenesis occurs during the first trimester of pregnancy culminating in the formation of hemangioblasts from intra-villous stem cells. Placental angiogenesis occurs subsequently, forming new blood vessels from existing ones. Angiogenesis also takes place at the fetomaternal interface, allowing essential spiral arteriole remodeling to establish the fetomaternal circulation. Vasculogenesis and angiogenesis in animal models and in humans have been studied in a wide variety of in vitro, physiological and pathological conditions, with a focus on the pro- and anti-angiogenic factors that control these processes. Recent studies revealed roles for new families of proteins, including direct participants such as the prokineticin family, and regulators of these processes such as the homeobox genes. This review summarizes recent advances in understanding the molecular mechanisms of actions of these families of proteins. Over the past decade, evidence suggests increased production of placental anti-angiogenic factors, as well as angiogenic factors are associated with fetal growth restriction (FGR) and preeclampsia (PE): the most threatening pathologies of human pregnancy with systemic vascular dysfunction. This review also reports novel clinical strategies targeting members of these family of proteins to treat PE and its consequent effects on the maternal vascular system.
Collapse
Affiliation(s)
- Nadia Alfaidy
- Unité 1036, Institut National de la Santé et de la Recherche Médicale, Grenoble, France.,Department of Biology, University of Grenoble Alpes, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
| | - Sophie Brouillet
- INSERM U1203, Department of Reproductive Biology, University of Montpellier, Montpellier, France
| | - Gayathri Rajaraman
- Faculty of Health and Biomedicine, First Year College, Victoria University, St. Albans, VIC, Australia
| | - Bill Kalionis
- Department of Maternal-Fetal Medicine, Obstetrics and Gynaecology, Pregnancy Research Centre, Royal Women's Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Pascale Hoffmann
- Unité 1036, Institut National de la Santé et de la Recherche Médicale, Grenoble, France.,Department of Biology, University of Grenoble Alpes, Grenoble, France.,Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
| | - Tiphaine Barjat
- Unité 1059, Saint-Etienne Hospital, Institut National de la Santé et de la Recherche Médicale, Saint-Étienne, France
| | - Mohamed Benharouga
- Unité Mixte de Recherche 5249, Laboratoire de Chimie et Biologie des Métaux, Centre National de la Recherche Scientifique (CNRS), Grenoble, France
| | - Padma Murthi
- Department of Maternal-Fetal Medicine, Obstetrics and Gynaecology, Pregnancy Research Centre, Royal Women's Hospital, The University of Melbourne, Parkville, VIC, Australia.,Department of Pharmacology, The Ritchie Centre, Monash Biomedicine Discovery Institute, Hudson Institute of Medical Research, Monash University, Clayton, VIC, Australia
| |
Collapse
|
49
|
Dayer G, Masoom ML, Togtema M, Zehbe I. Virus-Host Protein-Protein Interactions between Human Papillomavirus 16 E6 A1 and D2/D3 Sub-Lineages: Variances and Similarities. Int J Mol Sci 2020; 21:E7980. [PMID: 33121134 PMCID: PMC7663357 DOI: 10.3390/ijms21217980] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 10/23/2020] [Indexed: 01/07/2023] Open
Abstract
High-risk strains of human papillomavirus are causative agents for cervical and other mucosal cancers, with type 16 being the most frequent. Compared to the European Prototype (EP; A1), the Asian-American (AA; D2/D3) sub-lineage seems to have increased abilities to promote carcinogenesis. Here, we studied protein-protein interactions (PPIs) between host proteins and sub-lineages of the key transforming E6 protein. We transduced human keratinocyte with EP or AA E6 genes and co-immunoprecipitated E6 proteins along with interacting cellular proteins to detect virus-host binding partners. AAE6 and EPE6 may have unique PPIs with host cellular proteins, conferring gain or loss of function and resulting in varied abilities to promote carcinogenesis. Using liquid chromatography-mass spectrometry and stringent interactor selection criteria based on the number of peptides, we identified 25 candidates: 6 unique to AAE6 and EPE6, along with 13 E6 targets common to both. A novel approach based on pathway selection discovered 171 target proteins: 90 unique AAE6 and 61 unique EPE6 along with 20 common E6 targets. Interpretations were made using databases, such as UniProt, BioGRID, and Reactome. Detected E6 targets were differentially implicated in important hallmarks of cancer: deregulating Notch signaling, energetics and hypoxia, DNA replication and repair, and immune response.
Collapse
Affiliation(s)
- Guillem Dayer
- Biology Department, Lakehead University, Thunder Bay, ON P7B 5E1, Canada;
- Thunder Bay Regional Health Research Institute, Probe Development and Biomarker Exploration, Thunder Bay, ON P7B 6V4, Canada; (M.L.M.); (M.T.)
| | - Mehran L. Masoom
- Thunder Bay Regional Health Research Institute, Probe Development and Biomarker Exploration, Thunder Bay, ON P7B 6V4, Canada; (M.L.M.); (M.T.)
| | - Melissa Togtema
- Thunder Bay Regional Health Research Institute, Probe Development and Biomarker Exploration, Thunder Bay, ON P7B 6V4, Canada; (M.L.M.); (M.T.)
| | - Ingeborg Zehbe
- Biology Department, Lakehead University, Thunder Bay, ON P7B 5E1, Canada;
- Thunder Bay Regional Health Research Institute, Probe Development and Biomarker Exploration, Thunder Bay, ON P7B 6V4, Canada; (M.L.M.); (M.T.)
- Northern Ontario School of Medicine, Lakehead University, Thunder Bay, ON P7B 5E1, Canada
| |
Collapse
|
50
|
Lv Y, Du S, Huang X, Hao C. Follicular fluid estradiol is an improved predictor of in vitro fertilization/intracytoplasmic sperm injection and embryo transfer outcomes. Exp Ther Med 2020; 20:131. [PMID: 33082863 PMCID: PMC7557525 DOI: 10.3892/etm.2020.9256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 08/20/2019] [Indexed: 11/25/2022] Open
Abstract
The present study is a clinical trial analyzing follicular fluid. The current study aimed to assess whether a correlation exists among estradiol (E2), anti-Mullerian hormone (AMH) and prokineticin 1 (PROK1) levels in the follicular fluid. A total of 81 infertile patients (53 with primary infertility and 28 with secondary infertility) who received routine in vitro fertilization (IVF) and embryo transfer (ET) or intracytoplasmic sperm injection at Yuhuangding Hospital (Yantai, China) were included in the present study. On the day of egg retrieval, follicular puncture and follicular fluid extraction were performed on patients using double lumen needles under the guidance of a vaginal ultrasound. In 77 cases, follicular fluid was collected from the follicle with the largest diameter. A total of 53 cases underwent ET and subsequent pregnancy outcomes were traced. Concentrations of E2, AMH and PROK1 in the single follicular fluid specimens were determined. The concentration of E2 in follicular fluid from the largest follicles in absolute pregnancy group was significantly lower than that in absolute non-pregnancy group. The concentrations of PROK1 and AMH in follicular fluid from the largest follicles in absolute pregnancy group were not significantly different from those in absolute non-pregnancy group. The concentration of E2 was associated with the dosage of gonadotropin, but was not associated with age, AMH and PROK1 levels in follicular fluid, fertilization rate or number of usable blastocysts. The area under curve revealed that E2 level in the follicular fluid exhibited a low predictive value for pregnancy outcome. The present study demonstrated that E2 level is a better predictor for the outcome of IVF-ET than AMH or PROK1 levels in the follicular fluid.
Collapse
Affiliation(s)
- Yan Lv
- Clinical Medical College of Shandong University, Jinan, Shandong 250012, P.R. China.,Department of Gynecology and Reproductive Medicine, Jinan People's Hospital, Jinan, Shandong 271100, P.R. China
| | - Shengye Du
- Department of Obstetrics, Jinan People's Hospital, Jinan, Shandong 271100, P.R. China
| | - Xin Huang
- Reproduction Medical Center, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Cuifang Hao
- Reproduction Medical Center, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| |
Collapse
|