1
|
Luo JQ, Wang L, Liao ZQ, Lu BX, Luo CY, He HY, Ou Yang ZH, Duan SB, He SH, Wei AY, Zhang HB. Adipose stem cells ameliorate erectile dysfunction in diabetes mellitus rats by attenuating ferroptosis through NRP1 with SLC7A11 interaction. Free Radic Biol Med 2025; 232:40-55. [PMID: 40020883 DOI: 10.1016/j.freeradbiomed.2025.02.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Adipose stem cells (ADSCs) have garneVred increasing attention for their potential to treat diabetes mellitus erectile dysfunction (DMED), but the underlying molecular mechanisms remain unclear. The aim of this study was to identify and investigate the key cytokines and mechanisms by which ADSCs improve erectile function in DMED rats. METHODS We performed in vivo and in vitro assays, including rat erectile function assessment, cell co-culture, cytokine microarray screening and co-immunoprecipitation to investigate the role of ADSCs in improving erectile function in DMED rats. RESULTS Our analyses confirmed the occurrence of ferroptosis in the corpus cavernosum of DMED rats, while ADSCs treatment significantly restored erectile function and improved relevant indicators of ferroptosis. In vitro assays further indicated that corpus cavernosum smooth muscle cells (CCSMCs) co-cultured with ADSCs exhibited enhanced resistance to ferroptosis, with notably lower levels of cytoplasmic and lipid reactive oxygen species compared to the ferroptosis inducer Erastin-treated group. Mechanistic studies revealed that Neuropilin 1 (NRP1) may be a key molecule in ADSCs to improve erectile function in DMED rats. Furthermore, NRP1 in CCSMCs can interact with solute carrier family 7 member 11 (SLC7A11) to enhance the function of the glutamate-cysteine countertransport (Xc-) system and ferroptosis resistance in CCSMCs. CONCLUSION In conclusion, our findings indicate that NRP1 is a key molecule for ADSCs treatment to alleviate ferroptosis and improve erectile function in DMED rats, providing a promising target for DMED treatment and prognosis.
Collapse
Affiliation(s)
- Jun-Qi Luo
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Li Wang
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zi-Qi Liao
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Bing-Xin Lu
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Cai-Yu Luo
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Hai-Yang He
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Zhi-Han Ou Yang
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Song-Bo Duan
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Shu-Hua He
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China.
| | - An-Yang Wei
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China; Department of Urology, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, Guangdong, 511340, China.
| | - Hai-Bo Zhang
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
2
|
Bautista GM, Du Y, Matthews MJ, Flores AM, Kushnir NR, Sweeney NK, Nguyen NPN, Tokhtaeva E, Solorzano-Vargas RS, Lewis M, Stelzner M, He X, Dunn JCY, Martin MG. Smooth muscle cell Piezo1 depletion results in impaired contractile properties in murine small bowel. Commun Biol 2025; 8:448. [PMID: 40097724 PMCID: PMC11914552 DOI: 10.1038/s42003-025-07697-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
Piezo1 is a mechanosensitive cation channel expressed in intestinal muscularis cells (IMCs), including smooth muscle cells (SMCs), interstitial cells of Cajal, and Pdgfrα+ cells, which form the SIP syncytium, crucial for GI contractility. Here, we investigate the effects of SMC-specific Piezo1 deletion on small bowel function. Piezo1 depletion results in weight loss, delayed GI transit, muscularis thinning, and decreased SMCs. Ex vivo analyses demonstrated impaired contractile strength and tone, while in vitro studies using IMC co-cultures show dysrhythmic Ca2+ flux with decreased frequency. Imaging reveal that Piezo1 localizes intracellularly, thereby likely impacting Ca2+ signaling mechanisms modulated by Ca2 + -handling channels located on the sarcoplasmic reticulum and plasma membrane. Our findings suggest that Piezo1 in small bowel SMCs contributes to contractility by maintaining intracellular Ca2+ activity and subsequent signaling within the SIP syncytium. These findings provide new insights into the complex role of Piezo1 in small bowel SMCs and its implications for GI motility.
Collapse
Affiliation(s)
- Geoanna M Bautista
- Department of Pediatrics, Division of Neonatology, University of California Davis Children's Hospital, Sacramento, CA, 95817, USA
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Yingjie Du
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Michael J Matthews
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Allison M Flores
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Nicole R Kushnir
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Nicolle K Sweeney
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Nam Phuong N Nguyen
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Elmira Tokhtaeva
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - R S Solorzano-Vargas
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Michael Lewis
- Department of Pathology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Matthias Stelzner
- Department of Surgery, VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Ximin He
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - James C Y Dunn
- Division of Pediatric Surgery, Departments of Surgery and Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Martin G Martin
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA.
| |
Collapse
|
3
|
Zeng XL, Zhu LJ, Yang XD. Exploration of the complex origins of primary constipation. World J Clin Cases 2024; 12:5476-5482. [PMID: 39188609 PMCID: PMC11269998 DOI: 10.12998/wjcc.v12.i24.5476] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024] Open
Abstract
Constipation is a common gastrointestinal disorder characterized by infrequent bowel movements and difficulty in passing stools. It can significantly affect an individual's quality of life and overall well-being. Understanding the causes of constipation is important for its effective management and treatment. In this paper, we have reviewed the primary causes of constipation or functional constipation. Primary constipation is a bowel disorder associated with colonic or anorectal sensorimotor or neuromuscular dysfunction. As per the literature, it is multifactorial and involves factors such as decreased interstitial cells of Cajal, altered colonic motility, enteric nervous system dysfunction, intestinal flora disturbances, and psychological influences. Clinical symptoms include difficulty in defecation, decreased frequency of defecation, or a feeling of incomplete evacuation. A comprehensive evaluation and management of constipation require an interdisciplinary approach incorporating dietary modifications, lifestyle changes, pharmacotherapy, and psychological interventions. Further research is imperative to explain the intricate mechanisms underlying constipation and develop targeted therapies for improved patient outcomes.
Collapse
Affiliation(s)
- Xing-Lin Zeng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Lian-Jun Zhu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Xiang-Dong Yang
- Colorectal and Anal Surgery, Chengdu Anorectal Hospital, Chengdu 610015, Sichuan Province, China
| |
Collapse
|
4
|
Naidu AS, Wang CK, Rao P, Mancini F, Clemens RA, Wirakartakusumah A, Chiu HF, Yen CH, Porretta S, Mathai I, Naidu SAG. Precision nutrition to reset virus-induced human metabolic reprogramming and dysregulation (HMRD) in long-COVID. NPJ Sci Food 2024; 8:19. [PMID: 38555403 PMCID: PMC10981760 DOI: 10.1038/s41538-024-00261-2] [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: 10/12/2023] [Accepted: 03/15/2024] [Indexed: 04/02/2024] Open
Abstract
SARS-CoV-2, the etiological agent of COVID-19, is devoid of any metabolic capacity; therefore, it is critical for the viral pathogen to hijack host cellular metabolic machinery for its replication and propagation. This single-stranded RNA virus with a 29.9 kb genome encodes 14 open reading frames (ORFs) and initiates a plethora of virus-host protein-protein interactions in the human body. These extensive viral protein interactions with host-specific cellular targets could trigger severe human metabolic reprogramming/dysregulation (HMRD), a rewiring of sugar-, amino acid-, lipid-, and nucleotide-metabolism(s), as well as altered or impaired bioenergetics, immune dysfunction, and redox imbalance in the body. In the infectious process, the viral pathogen hijacks two major human receptors, angiotensin-converting enzyme (ACE)-2 and/or neuropilin (NRP)-1, for initial adhesion to cell surface; then utilizes two major host proteases, TMPRSS2 and/or furin, to gain cellular entry; and finally employs an endosomal enzyme, cathepsin L (CTSL) for fusogenic release of its viral genome. The virus-induced HMRD results in 5 possible infectious outcomes: asymptomatic, mild, moderate, severe to fatal episodes; while the symptomatic acute COVID-19 condition could manifest into 3 clinical phases: (i) hypoxia and hypoxemia (Warburg effect), (ii) hyperferritinemia ('cytokine storm'), and (iii) thrombocytosis (coagulopathy). The mean incubation period for COVID-19 onset was estimated to be 5.1 days, and most cases develop symptoms after 14 days. The mean viral clearance times were 24, 30, and 39 days for acute, severe, and ICU-admitted COVID-19 patients, respectively. However, about 25-70% of virus-free COVID-19 survivors continue to sustain virus-induced HMRD and exhibit a wide range of symptoms that are persistent, exacerbated, or new 'onset' clinical incidents, collectively termed as post-acute sequelae of COVID-19 (PASC) or long COVID. PASC patients experience several debilitating clinical condition(s) with >200 different and overlapping symptoms that may last for weeks to months. Chronic PASC is a cumulative outcome of at least 10 different HMRD-related pathophysiological mechanisms involving both virus-derived virulence factors and a multitude of innate host responses. Based on HMRD and virus-free clinical impairments of different human organs/systems, PASC patients can be categorized into 4 different clusters or sub-phenotypes: sub-phenotype-1 (33.8%) with cardiac and renal manifestations; sub-phenotype-2 (32.8%) with respiratory, sleep and anxiety disorders; sub-phenotype-3 (23.4%) with skeleto-muscular and nervous disorders; and sub-phenotype-4 (10.1%) with digestive and pulmonary dysfunctions. This narrative review elucidates the effects of viral hijack on host cellular machinery during SARS-CoV-2 infection, ensuing detrimental effect(s) of virus-induced HMRD on human metabolism, consequential symptomatic clinical implications, and damage to multiple organ systems; as well as chronic pathophysiological sequelae in virus-free PASC patients. We have also provided a few evidence-based, human randomized controlled trial (RCT)-tested, precision nutrients to reset HMRD for health recovery of PASC patients.
Collapse
Affiliation(s)
- A Satyanarayan Naidu
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA.
- N-terminus Research Laboratory, 232659 Via del Rio, Yorba Linda, CA, 92887, USA.
| | - Chin-Kun Wang
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- School of Nutrition, Chung Shan Medical University, 110, Section 1, Jianguo North Road, Taichung, 40201, Taiwan
| | - Pingfan Rao
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- College of Food and Bioengineering, Fujian Polytechnic Normal University, No.1, Campus New Village, Longjiang Street, Fuqing City, Fujian, China
| | - Fabrizio Mancini
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- President-Emeritus, Parker University, 2540 Walnut Hill Lane, Dallas, TX, 75229, USA
| | - Roger A Clemens
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- University of Southern California, Alfred E. Mann School of Pharmacy/D. K. Kim International Center for Regulatory & Quality Sciences, 1540 Alcazar St., CHP 140, Los Angeles, CA, 90089, USA
| | - Aman Wirakartakusumah
- International Union of Food Science and Technology (IUFoST), Guelph, ON, Canada
- IPMI International Business School Jakarta; South East Asian Food and Agriculture Science and Technology, IPB University, Bogor, Indonesia
| | - Hui-Fang Chiu
- Department of Chinese Medicine, Taichung Hospital, Ministry of Health & Well-being, Taichung, Taiwan
| | - Chi-Hua Yen
- Department of Family and Community Medicine, Chung Shan Medical University Hospital; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Sebastiano Porretta
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- President, Italian Association of Food Technology (AITA), Milan, Italy
- Experimental Station for the Food Preserving Industry, Department of Consumer Science, Viale Tanara 31/a, I-43121, Parma, Italy
| | - Issac Mathai
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- Soukya International Holistic Health Center, Whitefield, Bengaluru, India
| | - Sreus A G Naidu
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- N-terminus Research Laboratory, 232659 Via del Rio, Yorba Linda, CA, 92887, USA
| |
Collapse
|
5
|
Chen Z, Li YY, Liu X. Copper homeostasis and copper-induced cell death: Novel targeting for intervention in the pathogenesis of vascular aging. Biomed Pharmacother 2023; 169:115839. [PMID: 37976889 DOI: 10.1016/j.biopha.2023.115839] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/25/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023] Open
Abstract
Copper-induced cell death, also known as cuproptosis, is distinct from other types of cell death such as apoptosis, necrosis, and ferroptosis. It can trigger the accumulation of lethal reactive oxygen species, leading to the onset and progression of aging. The significant increases in copper ion levels in the aging populations confirm a close relationship between copper homeostasis and vascular aging. On the other hand, vascular aging is also closely related to the occurrence of various cardiovascular diseases throughout the aging process. However, the specific causes of vascular aging are not clear, and different living environments and stress patterns can lead to individualized vascular aging. By exploring the correlations between copper-induced cell death and vascular aging, we can gain a novel perspective on the pathogenesis of vascular aging and enhance the prognosis of atherosclerosis. This article aims to provide a comprehensive review of the impacts of copper homeostasis on vascular aging, including their effects on endothelial cells, smooth muscle cells, oxidative stress, ferroptosis, intestinal flora, and other related factors. Furthermore, we intend to discuss potential strategies involving cuproptosis and provide new insights for copper-related vascular aging.
Collapse
Affiliation(s)
- Zhuoying Chen
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China
| | - Yuan-Yuan Li
- Department of Nursing, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China.
| | - Xiangjie Liu
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430077, China.
| |
Collapse
|
6
|
Lambrinos G, Cristofaro V, Pelton K, Bigger-Allen A, Doyle C, Vasquez E, Bielenberg DR, Sullivan MP, Adam RM. Neuropilin 2 Is a Novel Regulator of Distal Colon Contractility. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1592-1603. [PMID: 35985479 PMCID: PMC9667714 DOI: 10.1016/j.ajpath.2022.07.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 06/05/2023]
Abstract
Appropriate coordination of smooth muscle contraction and relaxation is essential for normal colonic motility. The impact of perturbed motility ranges from moderate, in conditions such as colitis, to potentially fatal in the case of pseudo-obstruction. The mechanisms underlying aberrant motility and the extent to which they can be targeted pharmacologically are incompletely understood. This study identified colonic smooth muscle as a major site of expression of neuropilin 2 (Nrp2) in mice and humans. Mice with inducible smooth muscle-specific knockout of Nrp2 had an increase in evoked contraction of colonic rings in response to carbachol at 1 and 4 weeks following initiation of deletion. KCl-induced contractions were also increased at 4 weeks. Colonic motility was similarly enhanced, as evidenced by faster bead expulsion in Nrp2-deleted mice versus Nrp2-intact controls. In length-tension analysis of the distal colon, passive tension was similar in Nrp2-deficient and Nrp2-intact mice, but at low strains, active stiffness was greater in Nrp2-deficient animals. Consistent with the findings in conditional Nrp2 mice, Nrp2-null mice showed increased contractility in response to carbachol and KCl. Evaluation of selected proteins implicated in smooth muscle contraction revealed no significant differences in the level of α-smooth muscle actin, myosin light chain, calponin, or RhoA. Together, these findings identify Nrp2 as a novel regulator of colonic contractility that may be targetable in conditions characterized by dysmotility.
Collapse
Affiliation(s)
- George Lambrinos
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts
| | - Vivian Cristofaro
- Department of Surgery, Harvard Medical School, Boston, Massachusetts; Division of Urology, VA Boston Healthcare System, Boston, Massachusetts
| | - Kristine Pelton
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts
| | - Alexander Bigger-Allen
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts; Biological and Biomedical Sciences Program, Division of Medical Sciences, Harvard Medical School, Boston, Massachusetts
| | - Claire Doyle
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts
| | - Evalynn Vasquez
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts
| | - Diane R Bielenberg
- Department of Surgery, Harvard Medical School, Boston, Massachusetts; Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts
| | - Maryrose P Sullivan
- Department of Surgery, Harvard Medical School, Boston, Massachusetts; Division of Urology, VA Boston Healthcare System, Boston, Massachusetts.
| | - Rosalyn M Adam
- Urological Diseases Research Center, Boston Children's Hospital, Boston, Massachusetts; Department of Surgery, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
7
|
SARS CoV-2 detected in neonatal stool remote from maternal COVID-19 during pregnancy. Pediatr Res 2022; 93:1375-1382. [PMID: 35986143 PMCID: PMC9388973 DOI: 10.1038/s41390-022-02266-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 05/04/2022] [Accepted: 05/08/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND In utero transmission of SARS coronavirus 2 (SARS-CoV-2) has not been fully investigated. We investigated whether newborns of mothers with COVID-19 during pregnancy might harbor SARS-CoV-2 in the gastrointestinal tract. METHODS This cohort study investigated stool from 14 newborns born at 25-41 weeks admitted at delivery to our urban academic hospital whose mothers had COVID-19 during pregnancy. Eleven mothers had COVID-19 resolved more than 10 weeks before delivery. Newborn stool was evaluated for SARS-CoV-2 RNA, Spike protein, and induction of inflammatory cytokines interleukin-6 (IL-6) and interferon-γ (IFN-γ) in macrophages. RESULTS Despite negative SARS CoV-2 nasal PCRs from all newborns, viral RNAs and Spike protein were detected in the stool of 11 out of 14 newborns as early as the first day of life and increased over time in 6. Stool homogenates from all 14 newborns elicited elevated inflammatory IL-6 and IFN-γ from macrophages. Most newborns were clinically well except for one death from gestational autoimmune liver disease and another who developed necrotizing enterocolitis. CONCLUSIONS These findings suggest in utero transmission of SARS-CoV-2 and possible persistent intestinal viral reservoirs in the newborns. Further investigation is required to understand the mechanisms and their clinical implications. IMPACT SARS-CoV-2 RNAs or Spike protein was detected in the stool of 11 out of 14 preterm newborns born to mothers with resolved COVID-19 weeks prior to delivery despite negative newborn nasal PCR swabs. These novel findings suggest risk of in utero SARS-CoV-2 transmission to the fetal intestine during gestation. The presence of SARS-CoV-2 RNAs and Spike protein in the intestines of newborns may potentially impact the development of the gut microbiome and the immune system; the long-term health impact on the preterm infants should be further investigated.
Collapse
|
8
|
Sfera A, Osorio C, Zapata Martín del Campo CM, Pereida S, Maurer S, Maldonado JC, Kozlakidis Z. Endothelial Senescence and Chronic Fatigue Syndrome, a COVID-19 Based Hypothesis. Front Cell Neurosci 2021; 15:673217. [PMID: 34248502 PMCID: PMC8267916 DOI: 10.3389/fncel.2021.673217] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/25/2021] [Indexed: 12/14/2022] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome is a serious illness of unknown etiology, characterized by debilitating exhaustion, memory impairment, pain and sleep abnormalities. Viral infections are believed to initiate the pathogenesis of this syndrome although the definite proof remains elusive. With the unfolding of COVID-19 pandemic, the interest in this condition has resurfaced as excessive tiredness, a major complaint of patients infected with the SARS-CoV-2 virus, often lingers for a long time, resulting in disability, and poor life quality. In a previous article, we hypothesized that COVID-19-upregulated angiotensin II triggered premature endothelial cell senescence, disrupting the intestinal and blood brain barriers. Here, we hypothesize further that post-viral sequelae, including myalgic encephalomyelitis/chronic fatigue syndrome, are promoted by the gut microbes or toxin translocation from the gastrointestinal tract into other tissues, including the brain. This model is supported by the SARS-CoV-2 interaction with host proteins and bacterial lipopolysaccharide. Conversely, targeting microbial translocation and cellular senescence may ameliorate the symptoms of this disabling illness.
Collapse
Affiliation(s)
- Adonis Sfera
- Patton State Hospital, San Bernardino, CA, United States
| | | | | | | | - Steve Maurer
- Patton State Hospital, San Bernardino, CA, United States
| | - Jose Campo Maldonado
- Department of Internal Medicine, The University of Texas Rio Grande Valley, Edinburg, TX, United States
| | - Zisis Kozlakidis
- International Agency for Research on Cancer (IARC), Lyon, France
| |
Collapse
|
9
|
Devaux CA, Lagier JC, Raoult D. New Insights Into the Physiopathology of COVID-19: SARS-CoV-2-Associated Gastrointestinal Illness. Front Med (Lausanne) 2021; 8:640073. [PMID: 33681266 PMCID: PMC7930624 DOI: 10.3389/fmed.2021.640073] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/20/2021] [Indexed: 12/17/2022] Open
Abstract
Although SARS-CoV-2 is considered a lung-tropic virus that infects the respiratory tract through binding to the ACE2 cell-surface molecules present on alveolar lungs epithelial cells, gastrointestinal symptoms have been frequently reported in COVID-19 patients. What can be considered an apparent paradox is that these symptoms (e.g., diarrhea), sometimes precede the development of respiratory tract illness as if the breathing apparatus was not its first target during viral dissemination. Recently, evidence was reported that the gut is an active site of replication for SARS-CoV-2. This replication mainly occurs in mature enterocytes expressing the ACE2 viral receptor and TMPRSS4 protease. In this review we question how SARS-CoV-2 can cause intestinal disturbances, whether there are pneumocyte-tropic, enterocyte-tropic and/or dual tropic strains of SARS-CoV-2. We examine two major models: first, that of a virus directly causing damage locally (e.g., by inducing apoptosis of infected enterocytes); secondly, that of indirect effect of the virus (e.g., by inducing changes in the composition of the gut microbiota followed by the induction of an inflammatory process), and suggest that both situations probably occur simultaneously in COVID-19 patients. We eventually discuss the consequences of the virus replication in brush border of intestine on long-distance damages affecting other tissues/organs, particularly lungs.
Collapse
Affiliation(s)
- Christian A. Devaux
- Aix-Marseille University, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
- CNRS, Marseille, France
| | - Jean-Christophe Lagier
- Aix-Marseille University, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| | - Didier Raoult
- Aix-Marseille University, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| |
Collapse
|
10
|
Molecular Characterization of Constipation Disease as Novel Phenotypes in CRISPR-Cas9-Generated Leptin Knockout Mice with Obesity. Int J Mol Sci 2020; 21:ijms21249464. [PMID: 33322729 PMCID: PMC7763920 DOI: 10.3390/ijms21249464] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022] Open
Abstract
(1) Background: We characterized a novel animal model with obesity-induced constipation because constipation is rarely known in genetically engineered mice (GEM); (2) Methods: The changes in the constipation parameters and mechanisms were analyzed in CRISPR-Cas9-mediated leptin (Lep) knockout (KO) mice from eight to 24 weeks; (3) Results: Significant constipation phenotypes were observed in the Lep KO mice since 16 weeks old. These mice showed a significant decrease in the gastrointestinal motility, mucosal layer thickness and ability for mucin secretion as well as the abnormal ultrastructure of Lieberkühn crypts in the transverse colon. The density or function of the enteric neurons, intestinal Cajal cells (ICC), smooth muscle cells, and the concentration of gastrointestinal (GI) hormones for the GI motility were remarkably changed in Lep KO mice. The downstream signaling pathway of muscarinic acetylcholine receptors (mAChRs) were activated in Lep KO mice, while the expression of adipogenesis-regulating genes were alternatively reduced in the transverse colon of the same mice; (4) Conclusions: These results provide the first strong evidence that Lep KO mice can represent constipation successfully through dysregulation of the GI motility mediated by myenteric neurons, ICC, and smooth muscle cells in the transverse colon during an abnormal function of the lipid metabolism.
Collapse
|
11
|
The role of semaphorins in small vessels of the eye and brain. Pharmacol Res 2020; 160:105044. [PMID: 32590102 DOI: 10.1016/j.phrs.2020.105044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 12/20/2022]
Abstract
Small vessel diseases, such as ischemic retinopathy and cerebral small vessel disease (CSVD), are increasingly recognized in patients with diabetes, dementia and cerebrovascular disease. The mechanisms of small vessel diseases are poorly understood, but the latest studies suggest a role for semaphorins. Initially identified as axon guidance cues, semaphorins are mainly studied in neuronal morphogenesis, neural circuit assembly, and synapse assembly and refinement. In recent years, semaphorins have been found to play important roles in regulating vascular growth and development and in many pathophysiological processes, including atherosclerosis, angiogenesis after stroke and retinopathy. Growing evidence indicates that semaphorins affect the occurrence, perfusion and regression of both the macrovasculature and microvasculature by regulating the proliferation, apoptosis, migration, barrier function and inflammatory response of endothelial cells, vascular smooth muscle cells (VSMCs) and pericytes. In this review, we concentrate on the regulatory effects of semaphorins on the cell components of the vessel wall and their potential roles in microvascular diseases, especially in the retina and cerebral small vessel. Finally, we discuss potential molecular approaches in targeting semaphorins as therapies for microvascular disorders in the eye and brain.
Collapse
|
12
|
Mazzone A, Strege PR, Gibbons SJ, Alcaino C, Joshi V, Haak AJ, Tschumperlin DJ, Bernard CE, Cima RR, Larson DW, Chua HK, Graham RP, El Refaey M, Mohler PJ, Hayashi Y, Ordog T, Calder S, Du P, Farrugia G, Beyder A. microRNA overexpression in slow transit constipation leads to reduced Na V1.5 current and altered smooth muscle contractility. Gut 2020; 69:868-876. [PMID: 31757880 PMCID: PMC7147984 DOI: 10.1136/gutjnl-2019-318747] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 10/16/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study was designed to evaluate the roles of microRNAs (miRNAs) in slow transit constipation (STC). DESIGN All human tissue samples were from the muscularis externa of the colon. Expression of 372 miRNAs was examined in a discovery cohort of four patients with STC versus three age/sex-matched controls by a quantitative PCR array. Upregulated miRNAs were examined by quantitative reverse transcription qPCR (RT-qPCR) in a validation cohort of seven patients with STC and age/sex-matched controls. The effect of a highly differentially expressed miRNA on a custom human smooth muscle cell line was examined in vitro by RT-qPCR, electrophysiology, traction force microscopy, and ex vivo by lentiviral transduction in rat muscularis externa organotypic cultures. RESULTS The expression of 13 miRNAs was increased in STC samples. Of those miRNAs, four were predicted to target SCN5A, the gene that encodes the Na+ channel NaV1.5. The expression of SCN5A mRNA was decreased in STC samples. Let-7f significantly decreased Na+ current density in vitro in human smooth muscle cells. In rat muscularis externa organotypic cultures, overexpression of let-7f resulted in reduced frequency and amplitude of contraction. CONCLUSIONS A small group of miRNAs is upregulated in STC, and many of these miRNAs target the SCN5A-encoded Na+ channel NaV1.5. Within this set, a novel NaV1.5 regulator, let-7f, resulted in decreased NaV1.5 expression, current density and reduced motility of GI smooth muscle. These results suggest NaV1.5 and miRNAs as novel diagnostic and potential therapeutic targets in STC.
Collapse
Affiliation(s)
- Amelia Mazzone
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Peter R Strege
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Simon J Gibbons
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Constanza Alcaino
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Vikram Joshi
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrew J Haak
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Cheryl E Bernard
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Robert R Cima
- Department of Colon and Rectal Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - David W Larson
- Department of Colon and Rectal Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Heidi K Chua
- Department of Colon and Rectal Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Mona El Refaey
- Departments of Physiology and Cell Biology, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA,Department of Internal Medicine, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Peter J Mohler
- Departments of Physiology and Cell Biology, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Yujiro Hayashi
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamas Ordog
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Stefan Calder
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Peng Du
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Gianrico Farrugia
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA .,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Arthur Beyder
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA .,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
13
|
Nakane S, Umeda M, Kawashiri SY, Mukaino A, Ichinose K, Higuchi O, Maeda Y, Nakamura H, Matsuo H, Kawakami A. Detecting gastrointestinal manifestations in patients with systemic sclerosis using anti-gAChR antibodies. Arthritis Res Ther 2020; 22:32. [PMID: 32085768 PMCID: PMC7035754 DOI: 10.1186/s13075-020-2128-z] [Citation(s) in RCA: 15] [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: 11/19/2019] [Accepted: 02/12/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Patients with systemic sclerosis (SSc) complicated by gastrointestinal dysmotility are difficult to treat and have high mortality. To clarify the pathogenesis of gastrointestinal manifestations, we aimed to demonstrate the association among the clinical features of SSc, the serological markers, the autoantibodies against nicotinic acetylcholine receptor at autonomic ganglia (gAChR). METHODS Fifty patients were enrolled and divided into two groups according to the presence or absence of gastrointestinal manifestations, and the characteristics were analyzed between these two groups. We measured biomarkers and the autoantibodies against two gAChRα3 and β4 subunits to test sera samples. Furthermore, patients were classified based on the presence or absence of anti-gAChR autoantibodies, and their clinical features were compared. RESULTS In patients with SSc and gastrointestinal manifestations, digital ulcers were more frequent (p = 0.050) and VEGF expression was significantly higher (p = 0.038). Seven subjects with SSc were seropositive for α3 subunit, whereas one patient was seropositive for β4 subunit. The mean level of anti-gAChRα3 autoantibodies in SSc patients with gastrointestinal manifestations was significantly higher than that in SSc patients without gastrointestinal manifestations (p = 0.001). The group of patients with SSc and gAChR autoantibodies had significantly higher endostatin levels (p = 0.046). CONCLUSIONS This study is the first to demonstrate that clinical characteristics of SSc patients with seropositivity for gAChR autoantibodies. Patients with SSc have circulating autoantibodies against gAChR, which may contribute to gastrointestinal manifestations associated with this disease, suggesting that gAChR-mediated autonomic neurotransmission may provide a pathomechanism for gastrointestinal dysmotility in SSc.
Collapse
Affiliation(s)
- Shunya Nakane
- Department of Neuroimmunology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Clinical Research, Nagasaki Kawatana Medical Center, Nagasaki, Japan
- Department of Neurology, Nagasaki Kawatana Medical Center, Nagasaki, Japan
- Department of Molecular Neurology and Therapeutics, Kumamoto University Hospital, 1-1-1, Honjo, Chuouku, Kumamoto-shi, Kumamoto, 860-8556 Japan
| | - Masataka Umeda
- Department of Immunology and Rheumatology, Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Shin-ya Kawashiri
- Department of Immunology and Rheumatology, Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Akihiro Mukaino
- Department of Molecular Neurology and Therapeutics, Kumamoto University Hospital, 1-1-1, Honjo, Chuouku, Kumamoto-shi, Kumamoto, 860-8556 Japan
- Department of Neurology and Strokology, Nagasaki University Hospital, Nagasaki, Japan
| | - Kunihiro Ichinose
- Department of Immunology and Rheumatology, Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Osamu Higuchi
- Department of Clinical Research, Nagasaki Kawatana Medical Center, Nagasaki, Japan
| | - Yasuhiro Maeda
- Department of Neuroimmunology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Clinical Research, Nagasaki Kawatana Medical Center, Nagasaki, Japan
- Department of Neurology, Nagasaki Kawatana Medical Center, Nagasaki, Japan
| | - Hideki Nakamura
- Department of Immunology and Rheumatology, Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hidenori Matsuo
- Department of Neurology, Nagasaki Kawatana Medical Center, Nagasaki, Japan
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
14
|
Cipriani G, Terhaar ML, Eisenman ST, Ji S, Linden DR, Wright AM, Sha L, Ordog T, Szurszewski JH, Gibbons SJ, Farrugia G. Muscularis Propria Macrophages Alter the Proportion of Nitrergic but Not Cholinergic Gastric Myenteric Neurons. Cell Mol Gastroenterol Hepatol 2019; 7:689-691.e4. [PMID: 30711545 PMCID: PMC6477476 DOI: 10.1016/j.jcmgh.2019.01.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 12/13/2022]
Key Words
- bmp2, bone morphogenetic protein 2
- chat+, choline acetyltransferase+
- csf1, colony stimulating factor 1
- huc/d, embryonic lethal, abnormal vision, drosophila-like protein 3/4 antigen
- mpm, muscularis propria macrophage
- na, numerical aperture
- nos, nitric oxide synthase
- wt, wild-type
Collapse
Affiliation(s)
| | | | | | - Sihan Ji
- Enteric NeuroScience Program, Mayo Clinic, Rochester, Minnesota,Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - David R. Linden
- Enteric NeuroScience Program, Mayo Clinic, Rochester, Minnesota
| | - Alec M. Wright
- Enteric NeuroScience Program, Mayo Clinic, Rochester, Minnesota
| | - Lei Sha
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Tamas Ordog
- Enteric NeuroScience Program, Mayo Clinic, Rochester, Minnesota
| | | | - Simon J. Gibbons
- Enteric NeuroScience Program, Mayo Clinic, Rochester, Minnesota,Corresponding authors:
| | - Gianrico Farrugia
- Enteric NeuroScience Program, Mayo Clinic, Rochester, Minnesota,Corresponding authors:
| |
Collapse
|
15
|
Mahmoud M, Evans IM, Mehta V, Pellet-Many C, Paliashvili K, Zachary I. Smooth muscle cell-specific knockout of neuropilin-1 impairs postnatal lung development and pathological vascular smooth muscle cell accumulation. Am J Physiol Cell Physiol 2019; 316:C424-C433. [PMID: 30649916 PMCID: PMC6457104 DOI: 10.1152/ajpcell.00405.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuropilin 1 (NRP1) is important for neuronal and cardiovascular development due to its role in conveying class 3 semaphorin and vascular endothelial growth factor signaling, respectively. NRP1 is expressed in smooth muscle cells (SMCs) and mediates their migration and proliferation in cell culture and is implicated in pathological SMC remodeling in vivo. To address the importance of Nrp1 for SMC function during development, we generated conditional inducible Nrp1 SMC-specific knockout mice. Induction of early postnatal SMC-specific Nrp1 knockout led to pulmonary hemorrhage associated with defects in alveogenesis and revealed a specific requirement for Nrp1 in myofibroblast recruitment to the alveolar septae and PDGF-AA-induced migration in vitro. Furthermore, SMC-specific Nrp1 knockout inhibited PDGF-BB-stimulated SMC outgrowth ex vivo in aortic ring assays and reduced pathological arterial neointima formation in vivo. In contrast, we observed little significant effect of SMC-specific Nrp1 knockout on neonatal retinal vascularization. Our results point to a requirement of Nrp1 in vascular smooth muscle and myofibroblast function in vivo, which may have relevance for postnatal lung development and for pathologies characterized by excessive SMC and/or myofibroblast proliferation.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Centre for Cardiovascular Biology and Medicine, BHF Laboratories, Division of Medicine, University College London , London , United Kingdom
| | - Ian M Evans
- Centre for Cardiovascular Biology and Medicine, BHF Laboratories, Division of Medicine, University College London , London , United Kingdom
| | - Vedanta Mehta
- Centre for Cardiovascular Biology and Medicine, BHF Laboratories, Division of Medicine, University College London , London , United Kingdom
| | - Caroline Pellet-Many
- Centre for Cardiovascular Biology and Medicine, BHF Laboratories, Division of Medicine, University College London , London , United Kingdom
| | - Ketevan Paliashvili
- Centre for Cardiovascular Biology and Medicine, BHF Laboratories, Division of Medicine, University College London , London , United Kingdom
| | - Ian Zachary
- Centre for Cardiovascular Biology and Medicine, BHF Laboratories, Division of Medicine, University College London , London , United Kingdom
| |
Collapse
|
16
|
Finney AC, Orr AW. Guidance Molecules in Vascular Smooth Muscle. Front Physiol 2018; 9:1311. [PMID: 30283356 PMCID: PMC6157320 DOI: 10.3389/fphys.2018.01311] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/30/2018] [Indexed: 12/21/2022] Open
Abstract
Several highly conserved families of guidance molecules, including ephrins, Semaphorins, Netrins, and Slits, play conserved and distinct roles in tissue remodeling during tissue patterning and disease pathogenesis. Primarily, these guidance molecules function as either secreted or surface-bound ligands that interact with their receptors to activate a variety of downstream effects, including cell contractility, migration, adhesion, proliferation, and inflammation. Vascular smooth muscle cells, contractile cells comprising the medial layer of the vessel wall and deriving from the mural population, regulate vascular tone and blood pressure. While capillaries lack a medial layer of vascular smooth muscle, mural-derived pericytes contribute similarly to capillary tone to regulate blood flow in various tissues. Furthermore, pericyte coverage is critical in vascular development, as perturbations disrupt vascular permeability and viability. During cardiovascular disease, smooth muscle cells play a more dynamic role in which suppression of contractile markers, enhanced proliferation, and migration lead to the progression of aberrant vascular remodeling. Since many types of guidance molecules are expressed in vascular smooth muscle and pericytes, these may contribute to blood vessel formation and aberrant remodeling during vascular disease. While vascular development is a large focus of the existing literature, studies emerged to address post-developmental roles for guidance molecules in pathology and are of interest as novel therapeutic targets. In this review, we will discuss the roles of guidance molecules in vascular smooth muscle and pericyte function in development and disease.
Collapse
Affiliation(s)
- Alexandra Christine Finney
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States
| | - Anthony Wayne Orr
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States
- Department of Pathology and Translational Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States
| |
Collapse
|
17
|
Cipriani G, Gibbons SJ, Miller KE, Yang DS, Terhaar ML, Eisenman ST, Ördög T, Linden DR, Gajdos GB, Szurszewski JH, Farrugia G. Change in Populations of Macrophages Promotes Development of Delayed Gastric Emptying in Mice. Gastroenterology 2018; 154:2122-2136.e12. [PMID: 29501441 PMCID: PMC5985210 DOI: 10.1053/j.gastro.2018.02.027] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Muscularis propria macrophages lie close to cells that regulate gastrointestinal motor function, including interstitial cells of Cajal (ICC) and myenteric neurons. In animal models of diabetic gastroparesis, development of delayed gastric emptying has been associated with loss of macrophages that express cytoprotective markers and reduced networks of ICC. Mice with long-term diabetes and normal gastric emptying have macrophages that express anti-inflammatory markers and have normal gastric ICC. Mice homozygous for the osteopetrosis spontaneous mutation in the colony-stimulating factor 1 gene (Csf1op/op) do not have macrophages; when they are given streptozotocin to induce diabetes, they do not develop delayed gastric emptying. We investigated whether population of the gastric muscularis propria of diabetic Csf1op/op mice with macrophages is necessary to change gastric emptying, ICC, and myenteric neurons and investigated the macrophage-derived factors that determine whether diabetic mice do or do not develop delayed gastric emptying. METHODS Wild-type and Csf1op/op mice were given streptozotocin to induce diabetes. Some Csf1op/op mice were given daily intraperitoneal injections of CSF1 for 7 weeks; gastric tissues were collected and cellular distributions were analyzed by immunohistochemistry. CD45+, CD11b+, F4/80+ macrophages were dissociated from gastric muscularis propria, isolated by flow cytometry and analyzed by quantitative real-time polymerase chain reaction. Cultured gastric muscularis propria from Csf1op/op mice was exposed to medium that was conditioned by culture with bone marrow-derived macrophages from wild-type mice. RESULTS Gastric muscularis propria from Csf1op/op mice given CSF1 contained macrophages; 11 of 15 diabetic mice given CSF1 developed delayed gastric emptying and had damaged ICC. In non-diabetic Csf1op/op mice, administration of CSF1 reduced numbers of gastric myenteric neurons but did not affect the proportion of nitrergic neurons or ICC. In diabetic Csf1op/op mice given CSF1 that developed delayed gastric emptying, the proportion of nitrergic neurons was the same as in non-diabetic wild-type controls. Medium conditioned by macrophages previously exposed to oxidative injury caused damage to ICC in cultured gastric muscularis propria from Csf1op/op mice; neutralizing antibodies against IL6R or TNF prevented this damage to ICC. CD45+, CD11b+, and F4/80+ macrophages isolated from diabetic wild-type mice with delayed gastric emptying expressed higher levels of messenger RNAs encoding inflammatory markers (IL6 and inducible nitric oxide synthase) and lower levels of messenger RNAs encoding markers of anti-inflammatory cells (heme oxygenase 1, arginase 1, and FIZZ1) than macrophages isolated from diabetic mice with normal gastric emptying. CONCLUSIONS In studies of Csf1op/op and wild-type mice with diabetes, we found delayed gastric emptying to be associated with increased production of inflammatory factors, and reduced production of anti-inflammatory factors, by macrophages, leading to loss of ICC.
Collapse
|
18
|
Pollock CE, Sutherland HG, Maher BH, Lea RA, Haupt LM, Frith A, Anne MacGregor E, Griffiths LR. The NRP1 migraine risk variant shows evidence of association with menstrual migraine. J Headache Pain 2018; 19:31. [PMID: 29671086 PMCID: PMC5906416 DOI: 10.1186/s10194-018-0857-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/06/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND In 2016, a large meta-analysis brought the number of susceptibility loci for migraine to 38. While sub-type analysis for migraine without aura (MO) and migraine with aura (MA) found some loci showed specificity to MO, the study did not test the loci with respect to other subtypes of migraine. This study aimed to test the hypothesis that single nucleotide polymorphisms (SNPs) robustly associated with migraine are individually or collectively associated with menstrual migraine (MM). METHODS Genotyping of migraine susceptibility SNPs was conducted using the Agena MassARRAY platform on DNA samples from 235 women diagnosed with menstrual migraine as per International Classification for Headache Disorders II (ICHD-II) criteria and 140 controls. Alternative genotyping methods including restriction fragment length polymorphism, pyrosequencing and Sanger sequencing were used for validation. Statistical analysis was performed using PLINK and SPSS. RESULTS Genotypes of 34 SNPs were obtained and investigated for their potential association with menstrual migraine. Of these SNPs, rs2506142 located near the neuropilin 1 gene (NRP1), was found to be significantly associated with menstrual migraine (p = 0.003). Genomic risk scores were calculated for all 34 SNPs as well as a subset of 7 SNPs that were nearing individual significance. Overall, this analysis suggested these SNPs to be weakly predictive of MM, but of no prognostic or diagnostic value. CONCLUSIONS Our results suggest that NRP1 may be important in the etiology of MM. It also suggests some genetic commonality between common migraine subtypes (MA and MO) and MM. The identification of associated SNPs may be the starting point to a better understanding of how genetic factors may contribute to the menstrual migraine sub-type.
Collapse
Affiliation(s)
- Charmaine E. Pollock
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Heidi G. Sutherland
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Bridget H. Maher
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Rodney A. Lea
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Larisa M. Haupt
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | | | - E. Anne MacGregor
- Centre for Neuroscience & Trauma, Blizard Institute of Cell and Molecular Science, London, UK
| | - Lyn R. Griffiths
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
19
|
McGowan SE, McCoy DM. Neuropilin-1 and platelet-derived growth factor receptors cooperatively regulate intermediate filaments and mesenchymal cell migration during alveolar septation. Am J Physiol Lung Cell Mol Physiol 2018. [PMID: 29543041 DOI: 10.1152/ajplung.00511.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Generation of secondary alveolar septa occurs primarily after birth in humans and is complete in mice postnatally, when mechanical stresses vary as air space pressure oscillates. Alveolar mesenchymal cells deposit elastic fibers, which limit cell strain; although when the elastic fiber network is incomplete, this function is also served by the intracellular cytoskeleton. Intermediate filament proteins support deformation during cell division and migration, which occur during septal elongation. Because platelet-derived growth factor receptor-α (PDGFRα) signaling is essential for alveolar septation, we hypothesized that neuropilin-1 (NRP1) may link PDGFRα to cytoskeletal deformation. During cell migration, NRP1 links receptor tyrosine kinase signaling to cytoskeletal and focal adhesion remodeling. Therefore, we examined the consequences of nrp1 gene deletion in alveolar mesenchymal cells (myofibroblasts and pericytes). NRP1 depletion reduced the proportion of mesenchymal cells that contain nestin and desmin within the subpopulation that lacked PDGFRα but contained PDGFRβ. Desmin was reduced at alveolar entry rings, air spaces were enlarged, and surface area was reduced after NRP1 depletion. PDGFRα and NRP1 colocalized to membrane lipid rafts, which are known to contain Src kinase. NRP1 depletion reduced alveolar mesenchymal cell migration and PDGF-A-mediated activation of Src kinase, which may limit accumulation of desmin at septal tips (alveolar entry rings). Cooperation between NRP1 and PDGF signaling is required for secondary septation, and manipulation of NRP1 could promote alveolar regeneration without producing fibrosis.
Collapse
Affiliation(s)
- Stephen E McGowan
- Department of Veterans Affairs Research Service and Department of Internal Medicine, University of Iowa Carver College of Medicine , Iowa City, Iowa
| | - Diann M McCoy
- Department of Veterans Affairs Research Service and Department of Internal Medicine, University of Iowa Carver College of Medicine , Iowa City, Iowa
| |
Collapse
|
20
|
Human airway smooth muscle cell proliferation from asthmatics is negatively regulated by semaphorin3A. Oncotarget 2018; 7:80238-80251. [PMID: 27791986 PMCID: PMC5348316 DOI: 10.18632/oncotarget.12884] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 10/06/2016] [Indexed: 12/11/2022] Open
Abstract
Airway smooth muscle (ASM) hyperplasia is a key feature of airway remodeling in development of lung diseases such as asthma. Anomalous proliferation of ASM cells directly contributes to ASM hyperplasia. However, the molecular mechanisms controlling ASM cell proliferation are not completely understood. Semaphorins are versatile regulators of various cellular processes including cell growth and proliferation. The role of semaphorins in ASM cell proliferation has remained to be addressed. Here, we report that semaphorin 3A (Sema3A) receptor, neuropilin 1 (Nrp1), is expressed on human ASM cells (HASMC) isolated from healthy and asthmatic donors and treatment of these cells with exogenous Sema3A inhibits growth factor-induced proliferation. Sema3A inhibitory effect on HASMC proliferation is associated with decreased tyrosine phosphorylation of PDGFR, downregulation of Rac1 activation, STAT3 and GSK-3β phosphorylation. Bronchial sections from severe asthmatics displayed immunoreactivity of Nrp1, suggestive of functional contribution of Sema3A-Nrp1 axis in airway remodeling. Together, our data suggest Sema3A-Nrp1 signaling as a novel regulatory pathway of ASM hyperplasia.
Collapse
|
21
|
Lu YW, Lowery AM, Sun LY, Singer HA, Dai G, Adam AP, Vincent PA, Schwarz JJ. Endothelial Myocyte Enhancer Factor 2c Inhibits Migration of Smooth Muscle Cells Through Fenestrations in the Internal Elastic Lamina. Arterioscler Thromb Vasc Biol 2017; 37:1380-1390. [PMID: 28473437 DOI: 10.1161/atvbaha.117.309180] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/25/2017] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Laminar flow activates myocyte enhancer factor 2 (MEF2) transcription factors in vitro to induce expression of atheroprotective genes in the endothelium. Here we sought to establish the role of Mef2c in the vascular endothelium in vivo. APPROACH AND RESULTS To study endothelial Mef2c, we generated endothelial-specific deletion of Mef2c using Tie2-Cre or Cdh5-Cre-ERT2 and examined aortas and carotid arteries by en face immunofluorescence. We observed enhanced actin stress fiber formation in the Mef2c-deleted thoracic aortic endothelium (laminar flow region), similar to those observed in normal aortic inner curvature (disturbed flow region). Furthermore, Mef2c deletion resulted in the de novo formation of subendothelial intimal cells expressing markers of differentiated smooth muscle in the thoracic aortas and carotids. Lineage tracing showed that these cells were not of endothelial origin. To define early events in intimal development, we induced endothelial deletion of Mef2c and examined aortas at 4 and 12 weeks postinduction. The number of intimal cell clusters increased from 4 to 12 weeks, but the number of cells within a cluster peaked at 2 cells in both cases, suggesting ongoing migration but minimal proliferation. Moreover, we identified cells extending from the media through fenestrations in the internal elastic lamina into the intima, indicating transfenestral smooth muscle migration. Similar transfenestral migration was observed in wild-type carotid arteries ligated to induce neointimal formation. CONCLUSIONS These results indicate that endothelial Mef2c regulates the endothelial actin cytoskeleton and inhibits smooth muscle cell migration into the intima.
Collapse
Affiliation(s)
- Yao Wei Lu
- From the Department of Molecular and Cellular Physiology (Y.W.L., A.M.L., L.-Y.S., H.A.S., A.P.A., P.A.V., J.J.S.), and Department of Ophthalmology (A.P.A.), Albany Medical College, NY; Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY (G.D.); and Department of Bioengineering, Northeastern University, Boston, MA (G.D.)
| | - Anthony M Lowery
- From the Department of Molecular and Cellular Physiology (Y.W.L., A.M.L., L.-Y.S., H.A.S., A.P.A., P.A.V., J.J.S.), and Department of Ophthalmology (A.P.A.), Albany Medical College, NY; Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY (G.D.); and Department of Bioengineering, Northeastern University, Boston, MA (G.D.)
| | - Li-Yan Sun
- From the Department of Molecular and Cellular Physiology (Y.W.L., A.M.L., L.-Y.S., H.A.S., A.P.A., P.A.V., J.J.S.), and Department of Ophthalmology (A.P.A.), Albany Medical College, NY; Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY (G.D.); and Department of Bioengineering, Northeastern University, Boston, MA (G.D.)
| | - Harold A Singer
- From the Department of Molecular and Cellular Physiology (Y.W.L., A.M.L., L.-Y.S., H.A.S., A.P.A., P.A.V., J.J.S.), and Department of Ophthalmology (A.P.A.), Albany Medical College, NY; Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY (G.D.); and Department of Bioengineering, Northeastern University, Boston, MA (G.D.)
| | - Guohao Dai
- From the Department of Molecular and Cellular Physiology (Y.W.L., A.M.L., L.-Y.S., H.A.S., A.P.A., P.A.V., J.J.S.), and Department of Ophthalmology (A.P.A.), Albany Medical College, NY; Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY (G.D.); and Department of Bioengineering, Northeastern University, Boston, MA (G.D.)
| | - Alejandro P Adam
- From the Department of Molecular and Cellular Physiology (Y.W.L., A.M.L., L.-Y.S., H.A.S., A.P.A., P.A.V., J.J.S.), and Department of Ophthalmology (A.P.A.), Albany Medical College, NY; Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY (G.D.); and Department of Bioengineering, Northeastern University, Boston, MA (G.D.)
| | - Peter A Vincent
- From the Department of Molecular and Cellular Physiology (Y.W.L., A.M.L., L.-Y.S., H.A.S., A.P.A., P.A.V., J.J.S.), and Department of Ophthalmology (A.P.A.), Albany Medical College, NY; Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY (G.D.); and Department of Bioengineering, Northeastern University, Boston, MA (G.D.)
| | - John J Schwarz
- From the Department of Molecular and Cellular Physiology (Y.W.L., A.M.L., L.-Y.S., H.A.S., A.P.A., P.A.V., J.J.S.), and Department of Ophthalmology (A.P.A.), Albany Medical College, NY; Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY (G.D.); and Department of Bioengineering, Northeastern University, Boston, MA (G.D.).
| |
Collapse
|
22
|
The expanding role of neuropilin: regulation of transforming growth factor-β and platelet-derived growth factor signaling in the vasculature. Curr Opin Hematol 2016; 23:260-7. [PMID: 26849476 DOI: 10.1097/moh.0000000000000233] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Long recognized for its role in regulation of vascular endothelial growth factor signaling, neuropilin (Nrp)1 has emerged as a modulator of additional signaling pathways critical for vascular development and function. Here we review two novel functions of Nrp1 in blood vessels: regulation of transforming growth factor-β (TGFβ) signaling in endothelial cells and regulation of platelet-derived growth factor (PDGF) signaling in vascular smooth muscle cells. RECENT FINDINGS Novel mouse models demonstrate that Nrp1 fulfills vascular functions independent of vascular endothelial growth factor signaling. These include modulation of TGFβ-dependent inhibition of endothelial sprouting during developmental angiogenesis and PDGF signaling in vascular smooth muscle cells during development and disease. SUMMARY Broadening our understanding of how and where Nrp1 functions in the vasculature is critical for the development of targeted therapeutics for cancer and vascular diseases such as atherosclerosis and retinopathies.
Collapse
|
23
|
Wnuk M, Anderegg MA, Graber WA, Buergy R, Fuster DG, Djonov V. Neuropilin1 regulates glomerular function and basement membrane composition through pericytes in the mouse kidney. Kidney Int 2016; 91:868-879. [PMID: 27988210 DOI: 10.1016/j.kint.2016.10.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 12/28/2022]
Abstract
Neuropilin1 (Nrp1) is a co-receptor best known to regulate the development of endothelial cells and is a target of anticancer therapies. However, its role in other vascular cells including pericytes is emergent. The kidney is an organ with high pericyte density and cancer patients develop severe proteinuria following administration of NRP1B-neutralizing antibody combined with bevacizumab. Therefore, we investigated whether Nrp1 regulates glomerular capillary integrity after completion of renal development using two mouse models; tamoxifen-inducible NG2Cre to delete Nrp1 specifically in pericytes and administration of Nrp1-neutralizing antibodies. Specific Nrp1 deletion in pericytes did not affect pericyte number but mutant mice developed hematuria with glomerular basement membrane defects. Despite foot process effacement, albuminuria was absent and expression of podocyte proteins remained unchanged upon Nrp1 deletion. Additionally, these mice displayed dilation of the afferent arteriole and glomerular capillaries leading to glomerular hyperfiltration. Nidogen-1 mRNA was downregulated and collagen4α3 mRNA was upregulated with no significant effect on the expression of other basement membrane genes in the mutant mice. These features were phenocopied by treating wild-type mice with Nrp1-neutralizing antibodies. Thus, our results reveal a postdevelopmental role of Nrp1 in renal pericytes as an important regulator of glomerular basement membrane integrity. Furthermore, our study offers novel mechanistic insights into renal side effects of Nrp1 targeting cancer therapies.
Collapse
Affiliation(s)
- Monika Wnuk
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Manuel A Anderegg
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | | | - Regula Buergy
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Daniel G Fuster
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland; Division of Nephrology, Hypertension, and Clinical Pharmacology, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Bern, Switzerland.
| |
Collapse
|
24
|
Gormley P, Anttila V, Winsvold BS, Palta P, Esko T, Pers TH, Farh KH, Cuenca-Leon E, Muona M, Furlotte NA, Kurth T, Ingason A, McMahon G, Ligthart L, Terwindt GM, Kallela M, Freilinger TM, Ran C, Gordon SG, Stam AH, Steinberg S, Borck G, Koiranen M, Quaye L, Adams HHH, Lehtimäki T, Sarin AP, Wedenoja J, Hinds DA, Buring JE, Schürks M, Ridker PM, Hrafnsdottir MG, Stefansson H, Ring SM, Hottenga JJ, Penninx BWJH, Färkkilä M, Artto V, Kaunisto M, Vepsäläinen S, Malik R, Heath AC, Madden PAF, Martin NG, Montgomery GW, Kurki MI, Kals M, Mägi R, Pärn K, Hämäläinen E, Huang H, Byrnes AE, Franke L, Huang J, Stergiakouli E, Lee PH, Sandor C, Webber C, Cader Z, Muller-Myhsok B, Schreiber S, Meitinger T, Eriksson JG, Salomaa V, Heikkilä K, Loehrer E, Uitterlinden AG, Hofman A, van Duijn CM, Cherkas L, Pedersen LM, Stubhaug A, Nielsen CS, Männikkö M, Mihailov E, Milani L, Göbel H, Esserlind AL, Christensen AF, Hansen TF, Werge T, Kaprio J, Aromaa AJ, Raitakari O, Ikram MA, Spector T, Järvelin MR, Metspalu A, Kubisch C, Strachan DP, Ferrari MD, Belin AC, Dichgans M, Wessman M, van den Maagdenberg AMJM, Zwart JA, Boomsma DI, Smith GD, et alGormley P, Anttila V, Winsvold BS, Palta P, Esko T, Pers TH, Farh KH, Cuenca-Leon E, Muona M, Furlotte NA, Kurth T, Ingason A, McMahon G, Ligthart L, Terwindt GM, Kallela M, Freilinger TM, Ran C, Gordon SG, Stam AH, Steinberg S, Borck G, Koiranen M, Quaye L, Adams HHH, Lehtimäki T, Sarin AP, Wedenoja J, Hinds DA, Buring JE, Schürks M, Ridker PM, Hrafnsdottir MG, Stefansson H, Ring SM, Hottenga JJ, Penninx BWJH, Färkkilä M, Artto V, Kaunisto M, Vepsäläinen S, Malik R, Heath AC, Madden PAF, Martin NG, Montgomery GW, Kurki MI, Kals M, Mägi R, Pärn K, Hämäläinen E, Huang H, Byrnes AE, Franke L, Huang J, Stergiakouli E, Lee PH, Sandor C, Webber C, Cader Z, Muller-Myhsok B, Schreiber S, Meitinger T, Eriksson JG, Salomaa V, Heikkilä K, Loehrer E, Uitterlinden AG, Hofman A, van Duijn CM, Cherkas L, Pedersen LM, Stubhaug A, Nielsen CS, Männikkö M, Mihailov E, Milani L, Göbel H, Esserlind AL, Christensen AF, Hansen TF, Werge T, Kaprio J, Aromaa AJ, Raitakari O, Ikram MA, Spector T, Järvelin MR, Metspalu A, Kubisch C, Strachan DP, Ferrari MD, Belin AC, Dichgans M, Wessman M, van den Maagdenberg AMJM, Zwart JA, Boomsma DI, Smith GD, Stefansson K, Eriksson N, Daly MJ, Neale BM, Olesen J, Chasman DI, Nyholt DR, Palotie A. Meta-analysis of 375,000 individuals identifies 38 susceptibility loci for migraine. Nat Genet 2016; 48:856-66. [PMID: 27322543 DOI: 10.1038/ng.3598] [Show More Authors] [Citation(s) in RCA: 460] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 05/26/2016] [Indexed: 12/16/2022]
Abstract
Migraine is a debilitating neurological disorder affecting around one in seven people worldwide, but its molecular mechanisms remain poorly understood. There is some debate about whether migraine is a disease of vascular dysfunction or a result of neuronal dysfunction with secondary vascular changes. Genome-wide association (GWA) studies have thus far identified 13 independent loci associated with migraine. To identify new susceptibility loci, we carried out a genetic study of migraine on 59,674 affected subjects and 316,078 controls from 22 GWA studies. We identified 44 independent single-nucleotide polymorphisms (SNPs) significantly associated with migraine risk (P < 5 × 10(-8)) that mapped to 38 distinct genomic loci, including 28 loci not previously reported and a locus that to our knowledge is the first to be identified on chromosome X. In subsequent computational analyses, the identified loci showed enrichment for genes expressed in vascular and smooth muscle tissues, consistent with a predominant theory of migraine that highlights vascular etiologies.
Collapse
Affiliation(s)
- Padhraig Gormley
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Verneri Anttila
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Bendik S Winsvold
- FORMI, Oslo University Hospital, Oslo, Norway.,Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Priit Palta
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Tonu Esko
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Estonian Genome Center, University of Tartu, Tartu, Estonia.,Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Tune H Pers
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Kai-How Farh
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Illumina, San Diego, California, USA
| | - Ester Cuenca-Leon
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Pediatric Neurology, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Mikko Muona
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Folkhälsan Institute of Genetics, Helsinki, Finland.,Neuroscience Center, University of Helsinki, Helsinki, Finland.,Molecular Neurology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | | | - Tobias Kurth
- Institute of Public Health, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | - George McMahon
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Lannie Ligthart
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
| | - Gisela M Terwindt
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Mikko Kallela
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Tobias M Freilinger
- Department of Neurology and Epileptology, Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Caroline Ran
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Scott G Gordon
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Anine H Stam
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | | | - Guntram Borck
- Institute of Human Genetics, Ulm University, Ulm, Germany
| | - Markku Koiranen
- Center for Life Course Epidemiology and Systems Medicine, University of Oulu, Oulu, Finland
| | - Lydia Quaye
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Hieab H H Adams
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, School of Medicine, University of Tampere, Tampere, Finland
| | - Antti-Pekka Sarin
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Juho Wedenoja
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | | | - Julie E Buring
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Markus Schürks
- Department of Neurology, University Duisburg-Essen, Essen, Germany
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Susan M Ring
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, VU University Medical Centre, Amsterdam, the Netherlands
| | - Markus Färkkilä
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Ville Artto
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Mari Kaunisto
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Salli Vepsäläinen
- Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland
| | - Rainer Malik
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Andrew C Heath
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pamela A F Madden
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicholas G Martin
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Grant W Montgomery
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Mitja I Kurki
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Department of Neurosurgery, NeuroCenter, Kuopio University Hospital, Kuopio, Finland
| | - Mart Kals
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Kalle Pärn
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Eija Hämäläinen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Hailiang Huang
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Andrea E Byrnes
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jie Huang
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Evie Stergiakouli
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Phil H Lee
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Cynthia Sandor
- MRC Functional Genomics Unit, Department of Physiology, Anatomy &Genetics, Oxford University, Oxford, UK
| | - Caleb Webber
- MRC Functional Genomics Unit, Department of Physiology, Anatomy &Genetics, Oxford University, Oxford, UK
| | - Zameel Cader
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK.,Oxford Headache Centre, John Radcliffe Hospital, Oxford, UK
| | - Bertram Muller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, Christian Albrechts University, Kiel, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,National Institute for Health and Welfare, Helsinki, Finland
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Kauko Heikkilä
- Institute of Clinical Medicine, University of Helsinki, Helsinki, Finland
| | - Elizabeth Loehrer
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Lynn Cherkas
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | | | - Audun Stubhaug
- Department of Pain Management and Research, Oslo University Hospital, Oslo, Norway.,Medical Faculty, University of Oslo, Oslo, Norway
| | - Christopher S Nielsen
- Department of Pain Management and Research, Oslo University Hospital, Oslo, Norway.,Department of Ageing and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Minna Männikkö
- Center for Life Course Epidemiology and Systems Medicine, University of Oulu, Oulu, Finland
| | | | - Lili Milani
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | | | - Ann-Louise Esserlind
- Danish Headache Center, Department of Neurology, Rigshospitalet, Glostrup Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Anne Francke Christensen
- Danish Headache Center, Department of Neurology, Rigshospitalet, Glostrup Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Folkmann Hansen
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, University of Copenhagen, Roskilde, Denmark
| | - Thomas Werge
- Institute of Biological Psychiatry, MHC Sct. Hans, Mental Health Services Copenhagen, Copenhagen, Denmark.,Institute of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen, Copenhagen, Denmark.,iPSYCH-The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
| | | | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Department of Public Health, University of Helsinki, Helsinki, Finland.,Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Arpo J Aromaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Olli Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland.,Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Radiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Tim Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Marjo-Riitta Järvelin
- Center for Life Course Epidemiology and Systems Medicine, University of Oulu, Oulu, Finland.,Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPE) Centre for Environment and Health, School of Public Health, Imperial College London, London, UK.,Biocenter Oulu, University of Oulu, Oulu, Finland.,Unit of Primary Care, Oulu University Hospital, Oulu, Finland
| | | | - Christian Kubisch
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David P Strachan
- Population Health Research Institute, St George's, University of London, London, UK
| | - Michel D Ferrari
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Andrea C Belin
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Maija Wessman
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Arn M J M van den Maagdenberg
- Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands.,Department of Human Genetics, Leiden University Medical Centre, Leiden, the Netherlands
| | - John-Anker Zwart
- FORMI, Oslo University Hospital, Oslo, Norway.,Department of Neurology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
| | - George Davey Smith
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Kari Stefansson
- deCODE Genetics, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | - Mark J Daly
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin M Neale
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jes Olesen
- Danish Headache Center, Department of Neurology, Rigshospitalet, Glostrup Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Dale R Nyholt
- Statistical and Genomic Epidemiology Laboratory, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Aarno Palotie
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK.,Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Raimondi C, Brash JT, Fantin A, Ruhrberg C. NRP1 function and targeting in neurovascular development and eye disease. Prog Retin Eye Res 2016; 52:64-83. [PMID: 26923176 PMCID: PMC4854174 DOI: 10.1016/j.preteyeres.2016.02.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 12/19/2022]
Abstract
Neuropilin 1 (NRP1) is expressed by neurons, blood vessels, immune cells and many other cell types in the mammalian body and binds a range of structurally and functionally diverse extracellular ligands to modulate organ development and function. In recent years, several types of mouse knockout models have been developed that have provided useful tools for experimental investigation of NRP1 function, and a multitude of therapeutics targeting NRP1 have been designed, mostly with the view to explore them for cancer treatment. This review provides a general overview of current knowledge of the signalling pathways that are modulated by NRP1, with particular focus on neuronal and vascular roles in the brain and retina. This review will also discuss the potential of NRP1 inhibitors for the treatment for neovascular eye diseases.
Collapse
Affiliation(s)
- Claudio Raimondi
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - James T Brash
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK.
| |
Collapse
|
26
|
Scirocco A, Matarrese P, Carabotti M, Ascione B, Malorni W, Severi C. Cellular and Molecular Mechanisms of Phenotypic Switch in Gastrointestinal Smooth Muscle. J Cell Physiol 2016; 231:295-302. [PMID: 26206426 DOI: 10.1002/jcp.25105] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/21/2015] [Indexed: 10/16/2023]
Abstract
As a general rule, smooth muscle cells (SMC) are able to switch from a contractile phenotype to a less mature synthetic phenotype. This switch is accompanied by a loss of differentiation with decreased expression of contractile markers, increased proliferation as well as the synthesis and the release of several signaling molecules such as pro-inflammatory cytokines, chemotaxis-associated molecules, and growth factors. This SMC phenotypic plasticity has extensively been investigated in vascular diseases, but interest is also emerging in the field of gastroenterology. It has in fact been postulated that altered microenvironmental conditions, including the composition of microbiota, could trigger the remodeling of the enteric SMC, with phenotype changes and consequent alterations of contraction and impairment of gut motility. Several molecular actors participate in this phenotype remodeling. These include extracellular molecules such as cytokines and extracellular matrix proteins, as well as intracellular proteins, for example, transcription factors. Epigenetic control mechanisms and miRNA have also been suggested to participate. In this review key roles and actors of smooth muscle phenotypic switch, mainly in GI tissue, are described and discussed in the light of literature data available so far. J. Cell. Physiol. 231: 295-302, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Annunziata Scirocco
- Department of Internal Medicine and Medical Specialties, University Sapienza Rome, Rome, Italy
| | - Paola Matarrese
- Department of Drug Research and Evaluation, Istituto Superiore di Sanit, à, Rome, Italy
- Center of Metabolomics, Rome, Italy
| | - Marilia Carabotti
- Department of Internal Medicine and Medical Specialties, University Sapienza Rome, Rome, Italy
| | - Barbara Ascione
- Department of Drug Research and Evaluation, Istituto Superiore di Sanit, à, Rome, Italy
| | - Walter Malorni
- Department of Drug Research and Evaluation, Istituto Superiore di Sanit, à, Rome, Italy
- San Raffaele Pisana Institute, Rome, Italy
| | - Carola Severi
- Department of Internal Medicine and Medical Specialties, University Sapienza Rome, Rome, Italy
| |
Collapse
|
27
|
Calcium-Activated Potassium Channels: Potential Target for Cardiovascular Diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 104:233-261. [PMID: 27038376 DOI: 10.1016/bs.apcsb.2015.11.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Ca(2+)-activated K(+) channels (KCa) are classified into three subtypes: big conductance (BKCa), intermediate conductance (IKCa), and small conductance (SKCa) KCa channels. The three types of KCa channels have distinct physiological or pathological functions in cardiovascular system. BKCa channels are mainly expressed in vascular smooth muscle cells (VSMCs) and inner mitochondrial membrane of cardiomyocytes, activation of BKCa channels in these locations results in vasodilation and cardioprotection against cardiac ischemia. IKCa channels are expressed in VSMCs, endothelial cells, and cardiac fibroblasts and involved in vascular smooth muscle proliferation, migration, vessel dilation, and cardiac fibrosis. SKCa channels are widely expressed in nervous and cardiovascular system, and activation of SKCa channels mainly contributes membrane hyperpolarization. In this chapter, we summarize the physiological and pathological roles of the three types of KCa channels in cardiovascular system and put forward the possibility of KCa channels as potential target for cardiovascular diseases.
Collapse
|