1
|
Garton T, Smith MD, Kesharwani A, Gharagozloo M, Oh S, Na CH, Absinta M, Reich DS, Zack DJ, Calabresi PA. Myeloid lineage C3 induces reactive gliosis and neuronal stress during CNS inflammation. Nat Commun 2025; 16:3481. [PMID: 40216817 PMCID: PMC11992029 DOI: 10.1038/s41467-025-58708-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Complement component C3 mediates pathology in CNS neurodegenerative diseases. Here we use scRNAseq of sorted C3-reporter positive cells from mouse brain and optic nerve to characterize C3 producing glia in experimental autoimmune encephalomyelitis (EAE), a model in which peripheral immune cells infiltrate the CNS, causing reactive gliosis and neuro-axonal pathology. We find that C3 expression in the early inflammatory stage of EAE defines disease-associated glial subtypes characterized by increased expression of genes associated with mTOR activation and cell metabolism. This pro-inflammatory subtype is abrogated with genetic C3 depletion, a finding confirmed with proteomic analyses. In addition, early optic nerve axonal injury and retinal ganglion cell oxidative stress, but not loss of post-synaptic density protein 95, are ameliorated by selective deletion of C3 in myeloid cells. These data suggest that in addition to C3b opsonization of post synaptic proteins leading to neuronal demise, C3 activation is a contributor to reactive glia in the optic nerve.
Collapse
Affiliation(s)
- Thomas Garton
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Matthew D Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Ajay Kesharwani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Marjan Gharagozloo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Sungtaek Oh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Chan-Hyun Na
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Martina Absinta
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Reserach Hospital, Milan, Italy
| | - Daniel S Reich
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Donald J Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
2
|
Zelek WM, Bevan RJ, Nimmo J, Dewilde M, De Strooper B, Morgan BP. Brain-penetrant complement inhibition mitigates neurodegeneration in an Alzheimer's disease mouse model. Brain 2025; 148:941-954. [PMID: 39215579 PMCID: PMC11884734 DOI: 10.1093/brain/awae278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/10/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Complement activation is implicated in driving brain inflammation, self-cell damage and progression of injury in Alzheimer's disease and other neurodegenerative diseases. Here, we investigate the impact of brain delivery of a complement-blocking antibody on neurodegeneration in an Alzheimer's mouse model. We engineered a brain-penetrant recombinant antibody targeting the pro-inflammatory membrane attack complex. Systemic administration of this antibody in APPNL-G-F mice reduced brain levels of complement activation products, demonstrating successful brain entry and target engagement. Prolonged treatment decreased synapse loss, amyloid burden and brain inflammatory cytokine levels, concomitant with cognitive improvement compared to controls. These results underscore the potential of brain-penetrant complement-inhibiting drugs as promising therapeutics, targeting downstream of amyloid plaques in Alzheimer's disease.
Collapse
Affiliation(s)
- Wioleta M Zelek
- School of Medicine, UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF14 4XN, UK
| | - Ryan J Bevan
- School of Medicine, UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF14 4XN, UK
| | - Jacqui Nimmo
- School of Medicine, UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF14 4XN, UK
| | - Maarten Dewilde
- Therapeutic and Diagnostic Antibodies, Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven 3000, Belgium
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, Leuven 3000, Belgium
| | - Bart De Strooper
- Centre for Brain and Disease Research, KU Leuven and VIB Leuven, Leuven 3000, Belgium
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
| | - Bryan Paul Morgan
- School of Medicine, UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF14 4XN, UK
| |
Collapse
|
3
|
Keele GR, Dzieciatkowska M, Hay AM, Vincent M, O'Connor C, Stephenson D, Reisz JA, Nemkov T, Hansen KC, Page GP, Zimring JC, Churchill GA, D'Alessandro A. Genetic architecture of the red blood cell proteome in genetically diverse mice reveals central role of hemoglobin beta cysteine redox status in maintaining circulating glutathione pools. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640676. [PMID: 40093052 PMCID: PMC11908137 DOI: 10.1101/2025.02.27.640676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Red blood cells (RBCs) transport oxygen but accumulate oxidative damage over time, reducing function in vivo and during storage-critical for transfusions. To explore genetic influences on RBC resilience, we profiled proteins, metabolites, and lipids from fresh and stored RBCs obtained from 350 genetically diverse mice. Our analysis identified over 6,000 quantitative trait loci (QTL). Compared to other tissues, prevalence of trans genetic effects over cis reflects the absence of de novo protein synthesis in anucleated RBCs. QTL hotspots at Hbb, Hba, Mon1a, and storage-specific Steap3 linked ferroptosis to hemolysis. Proteasome components clustered at multiple loci, underscoring the importance of degrading oxidized proteins. Post-translational modifications (PTMs) mapped predominantly to hemoglobins, particularly cysteine residues. Loss of reactive C93 in humanized mice (HBB C93A) disrupted redox balance, affecting glutathione pools, protein glutathionylation, and redox PTMs. These findings highlight genetic regulation of RBC oxidation, with implications for transfusion biology and oxidative stress-dependent hemolytic disorders.
Collapse
|
4
|
Ovcinnikovs V, Dijkman K, Zom GG, Beurskens FJ, Trouw LA. Enhancing complement activation by therapeutic anti-tumor antibodies: Mechanisms, strategies, and engineering approaches. Semin Immunol 2025; 77:101922. [PMID: 39742715 DOI: 10.1016/j.smim.2024.101922] [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: 10/17/2024] [Revised: 12/06/2024] [Accepted: 12/07/2024] [Indexed: 01/04/2025]
Abstract
The complement system plays an integral role in both innate and adaptive immune responses. Beyond its protective function against infections, complement is also known to influence tumor immunity, where its activation can either promote tumor progression or mediate tumor cell destruction, depending on the context. One such context can be provided by antibodies, with their inherent capacity to activate the classical complement pathway. In recent years, our understanding of the mechanisms governing complement activation by IgG and IgM antibodies has expanded significantly. At the same time, preclinical and clinical studies on antibodies such as rituximab, ofatumumab, and daratumumab have provided evidence for the role of complement in therapeutic success, encouraging strategies to further enhance its activity. In this review we examine the main determinants of antibody-mediated complement activation, highlighting the importance of antibody subclass, affinity, valency, and geometry of antigen engagement. We summarize the evidence for complement involvement in anti-tumor activity and challenges of accurately estimating the extent of its contribution to therapeutic efficacy. Furthermore, we explore several engineering approaches designed to enhance complement activation, including increased Fc oligomerization and C1q affinity, bispecific C1q-recruiting antibodies, IgG subclass chimeras, as well as antibody and paratope combinations. Strategies targeting membrane-bound complement regulatory proteins to overcome tumor-associated complement inhibition are also discussed as a method to boost therapeutic efficacy. Finally, we highlight the potential of complement-dependent cellular cytotoxicity (CDCC) and complement-dependent cellular phagocytosis (CDCP) as effector mechanisms that warrant deeper investigation. By integrating advances in antibody and complement biology with insights from efforts to enhance complement activation in therapeutic antibodies, this review aims to provide a comprehensive framework of antibody design and engineering strategies that optimize complement activity for improved anti-tumor efficacy.
Collapse
Affiliation(s)
| | - Karin Dijkman
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | - Leendert A Trouw
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
5
|
Fernandes DC, Silva-de-França F, Pohl PC, Eto SF, Sardinha LR, Lambris JD, Tambourgi DV. Cp40-mediated complement C3 inhibition dampens inflammasome activation and inflammatory mediators storm induced by Bitis arietans venom. Int Immunopharmacol 2025; 147:113701. [PMID: 39809101 DOI: 10.1016/j.intimp.2024.113701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025]
Abstract
The complement system plays a crucial role in various pathophysiological conditions, including snake envenomation. In this study, we investigated the effects of Bitis arietans venom on the complement system using an ex vivo human whole blood model. Our findings demonstrate that B. arietans venom was able to activate the complement system, leading to a significant increase in the production of anaphylatoxins (C3a/C3a-desArg, C5a/C5a-desArg) and the soluble Terminal Complement Complex (sTCC). Inhibition of the C3 component by Cp40, a C3-C3b inhibitor, resulted in the reduction of C3a/C3a-desArg, C5a/C5a-desArg, and sTCC levels to baseline in venom-stimulated samples. Furthermore, treatment with Cp40 promoted a substantial decrease in the production of pro-inflammatory mediators, such as Prostaglandin E2 (PGE2), IL-8/CXCL8, MCP-1/CCL2, and MIG/CXCL9. To further elucidate the molecular mechanisms, we utilized the THP-1 cell line differentiated into M0 macrophages. Incubation of these macrophages with human plasma, from the human whole blood treated with B. arietans venom, resulted in the expression of the NLRP3 inflammasome and the production of IL-8 and IL-1β. Importantly, Cp40 was able to diminish the production of these cytokines, as well as the levels of ASC and caspase-1 proteins. In conclusion, our results indicate that the inhibition of the complement by Cp40 at C3/C3b level can modulate the inflammatory response and inflammasome activation induced by B. arietans venom. These findings suggest that complement inhibition may be a promising therapeutic approach for managing the inflammatory complications associated with this snake envenomation.
Collapse
Affiliation(s)
- Dayanne Carla Fernandes
- Immunochemistry Laboratory, Butantan Institute, São Paulo, SP, Brazil; Center of Toxins, Cell Signaling and Immune Response (CeTICS), CEPID, FAPESP, Brazil
| | - Felipe Silva-de-França
- Immunochemistry Laboratory, Butantan Institute, São Paulo, SP, Brazil; Center of Toxins, Cell Signaling and Immune Response (CeTICS), CEPID, FAPESP, Brazil
| | | | | | | | - John D Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Denise V Tambourgi
- Immunochemistry Laboratory, Butantan Institute, São Paulo, SP, Brazil; Center of Toxins, Cell Signaling and Immune Response (CeTICS), CEPID, FAPESP, Brazil.
| |
Collapse
|
6
|
Ruths L, Hengge J, Teixeira GQ, Haffner-Luntzer M, Ignatius A, Riegger J. Terminal complement complex deposition on chondrocytes promotes premature senescence in age- and trauma-related osteoarthritis. Front Immunol 2025; 15:1470907. [PMID: 39877352 PMCID: PMC11772281 DOI: 10.3389/fimmu.2024.1470907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
Background The complement system is locally activated after joint injuries and leads to the deposition of the terminal complement complex (TCC). Sublytic TCC deposition is associated with phenotypical alterations of human articular chondrocytes (hAC) and enhanced release of inflammatory cytokines. Chronic inflammation is a known driver of chondrosenescence in osteoarthritis (OA). Therefore, we investigated whether TCC deposition contributes to stress-induced premature senescence (SIPS) during aging in vivo and after ex vivo cartilage injury. Methods Femoral condyles of male 13-week-old and 72-week-old CD59-ko (higher TCC deposition), C6-deficient (insufficient TCC formation), and C57BL/6 (WT) mice were collected to assess age-related OA. Furthermore, macroscopically intact human and porcine cartilage explants were traumatized and cultured with/without 30% human serum (HS) to activate the complement system. Explants were additionally treated with clusterin (CLU, TCC inhibitor), N-acetylcysteine (NAC, antioxidant), Sarilumab (IL-6 receptor inhibitor), STAT3-IN-1 (STAT3 inhibitor), or IL-1 receptor antagonist (IL-1RA) in order to investigate the consequences of TCC deposition. Gene and protein expression of senescence-associated markers such as CDKN1A and CDKN2A was determined. Results In the murine aging model, CD59-ko mice developed after 72 weeks more severe OA compared to C6-deficient and WT mice. mRNA analysis revealed that the expression of Cdkn1a, Cdkn2a, Tp53, Il1b, and Il6 was significantly increased in the cartilage of CD59-ko mice. In human cartilage, trauma and subsequent stimulation with HS increased mRNA levels of CDKN1A, CDKN2A, and IL6, while inhibition of TCC formation by CLU reduced the expression. Antioxidative therapy with NAC had no anti-senescent effect. In porcine tissue, HS exposure and trauma had additive effects on the number of CDKN2A-positive cells, while Sarilumab, STAT-IN-1, and IL-1RA reduced CDKN2A expression by trend. Conclusion Our results demonstrate that complement activation and consequent TCC deposition is associated with chondrosenescence in age-related and trauma-induced OA. We provided evidence that the SIPS-like phenotype is more likely induced by TCC-mediated cytokine release rather than oxidative stress. Overall, targeting TCC formation could be a future approach to attenuate OA progression.
Collapse
Affiliation(s)
- Leonie Ruths
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University Medical Center, Ulm, Germany
| | - Jana Hengge
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University Medical Center, Ulm, Germany
| | - Graciosa Q. Teixeira
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
7
|
Silva de França F, de Oliveira Orsi R, Fernandes DC, Leonel TB, Tambourgi DV. Africanized honeybee venom ( Apis mellifera) promotes human complement activation split products storm. Front Immunol 2024; 15:1463471. [PMID: 39606222 PMCID: PMC11598452 DOI: 10.3389/fimmu.2024.1463471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/14/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction Complement activation split products are signatures of many immunopathological disorders. Among the laboratory findings observed in these diseases, a reduction in the level of circulating intact complement components can be mentioned, and this change has also been detected in envenomation by multiple Africanized honeybee (Apis mellifera) stings. Although envenomation by these animals elicits diverse life-threatening reactions, the capacity of bee venom (AmV) to activate the human complement system remains elusive. Methods and findings By coupling immunochemical and functional approaches, it was observed that AmV strongly consumes components of the alternative pathway (AP) of the complement system in normal human serum (NHS). Additionally, AmV interfered with classical (CP) and lectin pathways (LP) activities. In parallel, a high increase in Ba fragment levels was detected, suggesting that the changes in AP activity were due to its activation. Furthermore, an increase in the level of the C1s-C1INH complex and a decrease in the physiological level of MASP1-C1INH suggested that CP and LP were also activated in the presence of AmV. Strikingly, NHS exposed to increasing AmV concentrations varying from 5 to 1000 µg/mL presented a high generation of C3a, C4a and C5a anaphylatoxins, and sC5b-9 complexes assembly, thus reinforcing that AmV triggers complement activation. Conclusion These results show that AmV is a strong complement activator. This activation presents a mixed profile, with a predominance of AP activation. This suggests that complement split products can play important roles in the envenomation by Africanized honeybee, as they could induce diverse immunopathological events observed in patients and may also dictate patient clinical prognosis.
Collapse
Affiliation(s)
- Felipe Silva de França
- Immunochemistry Laboratory, Butantan Institute, São Paulo, Brazil
- Center of Toxins, Cell Signaling and Immune Response (CeTICS) – CEPID - FAPESP, São Paulo, Brazil
| | - Ricardo de Oliveira Orsi
- Center of Education, Science and Technology in Rational Beekeeping (NECTAR), College of Veterinary Medicine and Animal Sciences, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Dayanne Carla Fernandes
- Immunochemistry Laboratory, Butantan Institute, São Paulo, Brazil
- Center of Toxins, Cell Signaling and Immune Response (CeTICS) – CEPID - FAPESP, São Paulo, Brazil
| | - Thyago Bispo Leonel
- Immunochemistry Laboratory, Butantan Institute, São Paulo, Brazil
- Center of Toxins, Cell Signaling and Immune Response (CeTICS) – CEPID - FAPESP, São Paulo, Brazil
| | - Denise V. Tambourgi
- Immunochemistry Laboratory, Butantan Institute, São Paulo, Brazil
- Center of Toxins, Cell Signaling and Immune Response (CeTICS) – CEPID - FAPESP, São Paulo, Brazil
| |
Collapse
|
8
|
Murayama MA. Complement C3 deficient mice show more severe imiquimod-induced psoriasiform dermatitis than wild-type mice regardless of the commensal microbiota. Exp Anim 2024; 73:458-467. [PMID: 38945882 PMCID: PMC11534491 DOI: 10.1538/expanim.24-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024] Open
Abstract
The complement active product, C3a, and the receptor C3aR comprise an axis that exerts various biological functions, such as protection against infection. C3a is highly expressed in the inflamed skin and blood from patients with psoriasiform dermatitis. However, the role of the C3a/C3aR axis in psoriasiform dermatitis remains unclear because conflicting results using C3-/- mice have been published. In this study, to elucidate the contribution of commensal microbiota in C3-/- and wild-type (WT) mice were subjected to imiquimod-induced psoriasiform dermatitis under different housing conditions. C3-/- mice showed increased epidermal thickness and keratinocyte proliferation markers in the inflamed ear compared to WT mice upon treatment with IMQ. These inflamed phenotypes were observed in both cohoused and separately housed conditions, and antibiotic treatment did not abolish the aggravation of IMQ-induced psoriasiform dermatitis in C3-/- mice. These results suggested that the difference of commensal microbiota is not important for the C3-involved psoriasiform dermatitis. Keratinocyte hyperproliferation is a major feature of the inflamed skin in patients with psoriasiform dermatitis. In vitro experiments showed that C3a and C3aR agonists inhibited keratinocyte proliferation, which was abolished by introduction of a C3aR antagonist. Collectively, these results suggest that the C3a/C3aR axis plays a critical role in psoriasiform dermatitis development by inhibiting keratinocyte proliferation, regardless of the regulation of the commensal microbiota.
Collapse
Affiliation(s)
- Masanori A Murayama
- Department of Animal Models for Human Diseases, Institute of Biomedical Science, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
9
|
Ohta N, Matsuzaki T, Nakai M, Tabata Y, Nimura K. Combining mRNA with PBS and calcium ions improves the efficiency of the transfection of mRNA into tumors. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102273. [PMID: 39184192 PMCID: PMC11342175 DOI: 10.1016/j.omtn.2024.102273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/13/2024] [Indexed: 08/27/2024]
Abstract
mRNA is a promising modality for expressing a protein in vivo. Drug delivery systems are required for the efficient transfection of mRNA into cells. In this study, we evaluated several drug delivery systems for transfecting mRNA into tumors. A lipid nanoparticle delivered mRNA to the draining lymph nodes and liver, even by intratumoral injection. A liposome-based system did not consistently provide mRNA for different types of tumor cells. We found that PBS introduced mRNA into several tumors, and calcium ions enhanced the efficiency, particularly in male mice. The circular dichroism spectrometer suggested a structural change in mRNA in PBS. Transmission electron microscopy revealed that calcium ions promoted the formation of mRNA nanoparticles in PBS. Transfection of mRNAs coding OX40-ligand, interleukin (IL)-36γ, and IL-23 by PBS + calcium ions attenuated tumor growth. Our results indicate that combining PBS with calcium ions promotes the transfection of mRNA into tumors. These data provide information for the development of methods for transfection of mRNA for cancer therapy.
Collapse
Affiliation(s)
- Noriko Ohta
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
- Division of Gene Therapy Science, Gunma University Initiative for Advanced Research, Gunma University, Maebashi, Gunma 371-8511, Japan
| | - Takashi Matsuzaki
- Department of DDS Pharmaceutical Development, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Masayoshi Nakai
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yasuhiko Tabata
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Keisuke Nimura
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
- Division of Gene Therapy Science, Gunma University Initiative for Advanced Research, Gunma University, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
10
|
El-Attar NA, El-Sawi MR, El-Shabasy EA. The synergistic effect of Ficus carica nanoparticles and Praziquantel on mice infected by Schistosoma mansoni cercariae. Sci Rep 2024; 14:18944. [PMID: 39147839 PMCID: PMC11327331 DOI: 10.1038/s41598-024-68957-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/30/2024] [Indexed: 08/17/2024] Open
Abstract
Bilharzia is a parasitic flatworm that causes schistosomiasis, a neglected tropical illness worldwide. Praziquantel (PZQ) is a commercial single treatment of schistosomiasis so alternative drugs are needed to get rid of its side effects on the liver. The current study aimed to estimate the effective role of Ficus carica nanoparticles (Fc-NPCs), silver nanoparticles (Ag-NPCs) and Ficus carica nanoparticles loaded on silver nanoparticles (Fc-Ag NPCs) on C57BL/6 black female mice infected by Schistosoma mansoni and treated with PZQ treatment. It was proved that schistosomiasis causes liver damage in addition to the PZQ is ineffective as an anti-schistosomiasis; it is recorded in the infected mice group and PZQ treated group as in liver function tests, oxidative stress markers & anti-oxidants, pro-inflammatory markers, pro-apoptotic and anti-apoptotic markers also in liver cells' DNA damage. The amelioration in all tested parameters has been clarified in nanoparticle-protected mice groups. The Fc-Ag NPCs + PZQ group recorded the best preemptive effects as anti-schistosomiasis. Fc-NPCs, Ag-NPCs and Fc-Ag NPCs could antagonize PZQ effects that were observed in amelioration of all tested parameters. The study showed the phytochemicals' nanoparticles groups have an ameliorated effect on the health of infected mice.
Collapse
Affiliation(s)
- Naira A El-Attar
- Zoology Department, Faculty of Science, Mansoura University, Mansoura City, Egypt.
| | - Mamdouh R El-Sawi
- Zoology Department, Faculty of Science, Mansoura University, Mansoura City, Egypt
| | - Eman A El-Shabasy
- Zoology Department, Faculty of Science, Mansoura University, Mansoura City, Egypt
| |
Collapse
|
11
|
Aumailley L, Dubois MJ, Marette A, Lebel M. Integrated liver and serum proteomics uncover sexual dimorphism and alteration of several immune response proteins in an aging Werner syndrome mouse model. Aging (Albany NY) 2024; 16:8417-8445. [PMID: 38795389 PMCID: PMC11164518 DOI: 10.18632/aging.205866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 03/18/2024] [Indexed: 05/27/2024]
Abstract
Werner syndrome (WS) is a progeroid disorder caused by mutations in a protein containing both a DNA exonuclease and DNA helicase domains. Previous studies indicated that males lacking the helicase domain of the Wrn protein orthologue exhibited hepatic transcriptomic and metabolic alterations. In this study, we used a label-free liquid chromatography-tandem mass spectrometry approach to uncover proteins abundance associated with specific biological processes that differed depending on the age (four or ten months) and/or the genotype (wild type or Wrn mutant) in the serum and liver of mice. Principal component analysis of the proteomic data from both serum and hepatic tissue revealed a sexual dimorphism regardless of the age and the genotype of the mice. Moreover, although all Wrn mutant mice exhibited fatty liver by the age of ten months, a significant age and genotype dependent enrichment of proteins involved in lipid and fatty acid metabolic processes were uncovered only in males. Also, a genotype dependent increase in serum oxidant detoxification processes was observed in the serum of Wrn mutant males. Despite these sexual differences, several aspects of the immune system were affected in both females and males. Finally, an increase of specific immunoglobulin molecules was common in the liver and serum of both older Wrn mutant females and males. Such results suggest that specific immunoglobulin variants maybe associated with fatty liver progression in WS.
Collapse
Affiliation(s)
- Lucie Aumailley
- Centre de recherche du CHU de Québec, Faculty of Medicine, Université Laval, Québec City G1V 4G2, Canada
| | - Marie Julie Dubois
- Quebec Heart and Lung Institute, Faculty of Medicine, Université Laval, Québec City G1V 0A6, Canada
| | - André Marette
- Quebec Heart and Lung Institute, Faculty of Medicine, Université Laval, Québec City G1V 0A6, Canada
| | - Michel Lebel
- Centre de recherche du CHU de Québec, Faculty of Medicine, Université Laval, Québec City G1V 4G2, Canada
| |
Collapse
|
12
|
Davies ML, Biryukov SS, Rill NO, Klimko CP, Hunter M, Dankmeyer JL, Miller JA, Shoe JL, Mlynek KD, Talyansky Y, Toothman RG, Qiu J, Bozue JA, Cote CK. Sex differences in immune protection in mice conferred by heterologous vaccines for pneumonic plague. Front Immunol 2024; 15:1397579. [PMID: 38835755 PMCID: PMC11148226 DOI: 10.3389/fimmu.2024.1397579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/25/2024] [Indexed: 06/06/2024] Open
Abstract
Background Yersinia pestis is the etiological agent of plague, which can manifest as bubonic, septicemic, and/or pneumonic disease. Plague is a severe and rapidly progressing illness that can only be successfully treated with antibiotics initiated early after infection. There are no FDA-approved vaccines for plague, and some vaccine candidates may be less effective against pneumonic plague than bubonic plague. Y. pestis is not known to impact males and females differently in mechanisms of pathogenesis or severity of infection. However, one previous study reported sex-biased vaccine effectiveness after intranasal Y. pestis challenge. As part of developing a safe and effective vaccine, it is essential that potential sex differences are characterized. Methods In this study we evaluated novel vaccines in male and female BALB/c mice using a heterologous prime-boost approach and monitored survival, bacterial load in organs, and immunological correlates. Our vaccine strategy consisted of two subcutaneous immunizations, followed by challenge with aerosolized virulent nonencapsulated Y. pestis. Mice were immunized with a combination of live Y. pestis pgm- pPst-Δcaf1, live Y. pestis pgm- pPst-Δcaf1/ΔyopD, or recombinant F1-V (rF1-V) combined with adjuvants. Results The most effective vaccine regimen was initial priming with rF1-V, followed by boost with either of the live attenuated strains. However, this and other strategies were more protective in female mice. Males had higher bacterial burden and differing patterns of cytokine expression and serum antibody titers. Male mice did not demonstrate synergy between vaccination and antibiotic treatment as repeatedly observed in female mice. Conclusions This study provides new knowledge about heterologous vaccine strategies, sex differences in plague-vaccine efficacy, and the immunological factors that differ between male and female mice.
Collapse
Affiliation(s)
- Michael L Davies
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Sergei S Biryukov
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Nathaniel O Rill
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Christopher P Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Melissa Hunter
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Jennifer L Dankmeyer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Jeremy A Miller
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Jennifer L Shoe
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Kevin D Mlynek
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Yuli Talyansky
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Ronald G Toothman
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Ju Qiu
- Regulated Research Administration: Biostatistics Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Joel A Bozue
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| | - Christopher K Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, United States
| |
Collapse
|
13
|
Zelek WM, Bevan RJ, Morgan BP. Targeting terminal pathway reduces brain complement activation, amyloid load and synapse loss, and improves cognition in a mouse model of dementia. Brain Behav Immun 2024; 118:355-363. [PMID: 38485063 DOI: 10.1016/j.bbi.2024.03.017] [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: 12/05/2023] [Revised: 02/28/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024] Open
Abstract
Complement is dysregulated in the brain in Alzheimer's Disease and in mouse models of Alzheimer's disease. Each of the complement derived effectors, opsonins, anaphylatoxins and membrane attack complex (MAC), have been implicated as drivers of disease but their relative contributions remain unclarified. Here we have focussed on the MAC, a lytic and pro-inflammatory effector, in the AppNL-G-F mouse amyloidopathy model. To test the role of MAC, we back-crossed to generate AppNL-G-F mice deficient in C7, an essential MAC component. C7 deficiency ablated MAC formation, reduced synapse loss and amyloid load and improved cognition compared to complement-sufficient AppNL-G-F mice at 8-10 months age. Adding back C7 caused increased MAC formation in brain and an acute loss of synapses in C7-deficient AppNL-G-F mice. To explore whether C7 was a viable therapeutic target, a C7-blocking monoclonal antibody was administered systemically for one month in AppNL-G-F mice aged 8-9 months. Treatment reduced brain MAC and amyloid deposition, increased synapse density and improved cognitive performance compared to isotype control-treated AppNL-G-F mice. The findings implicate MAC as a driver of pathology and highlight the potential for complement inhibition at the level of MAC as a therapy in Alzheimer's disease.
Collapse
Affiliation(s)
- Wioleta M Zelek
- UK Dementia Research Institute Cardiff and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, Wales CF14 4XN, United Kingdom.
| | - Ryan J Bevan
- UK Dementia Research Institute Cardiff and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, Wales CF14 4XN, United Kingdom
| | - Bryan Paul Morgan
- UK Dementia Research Institute Cardiff and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, Wales CF14 4XN, United Kingdom.
| |
Collapse
|
14
|
Kelkar NS, Goldberg BS, Dufloo J, Bruel T, Schwartz O, Hessell AJ, Ackerman ME. Sex- and species-associated differences in complement-mediated immunity in humans and rhesus macaques. mBio 2024; 15:e0028224. [PMID: 38385704 PMCID: PMC10936177 DOI: 10.1128/mbio.00282-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/23/2024] Open
Abstract
The complement system can be viewed as a "moderator" of innate immunity, "instructor" of humoral immunity, and "regulator" of adaptive immunity. While sex is known to affect humoral and cellular immune systems, its impact on complement in humans and rhesus macaques, a commonly used non-human primate model system, has not been well studied. To address this knowledge gap, we analyzed serum samples from 90 humans and 72 rhesus macaques for the abundance and activity of the complement system components. While sequences of cascade proteins were highly conserved, dramatically different levels were observed between species. Whereas the low levels detected in rhesus samples raised questions about the suitability of the test for use with macaque samples, differences in levels of complement proteins were observed in male and female humans. Levels of total and antibody-dependent deposition of C1q and C3b on a glycosylated antigen differed between humans and rhesus, suggesting differential recognition of glycans and balance between classical and alternative activation pathways. Functional differences in complement-mediated lysis of antibody-sensitized cells were observed in multiple assays and showed that human females frequently exhibited higher lytic activity than human males or rhesus macaques, which typically did not exhibit such sex-associated differences. Other differences between species and sexes were observed in more narrow contexts-for only certain antibodies, antigens, or assays. Collectively, these results expand knowledge of sex-associated differences in the complement system in humans, identifying differences absent from rhesus macaques.IMPORTANCEThe complement system is a critical part of host defense to many bacterial, fungal, and viral infections. In parallel, rich epidemiological, clinical, and biomedical research evidence demonstrates that sex is an important biological variable in immunity, and many sex-specific differences in immune system are intimately tied with disease outcomes. This study focuses on the intersection of these two factors to define the impact of sex on complement pathway components and activities. This work expands our knowledge of sex-associated differences in the complement system in humans and also identifies the differences that appear to be absent in rhesus macaques, a popular non-human primate model. Whereas differences between species suggest potential limitations in the ability of macaque model to recapitulate human biology, knowledge of sex-based differences in humans has the potential to inform clinical research and practice.
Collapse
Affiliation(s)
- Natasha S. Kelkar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, New Hampshire, USA
| | | | - Jérémy Dufloo
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France
| | - Timothée Bruel
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France
- Vaccine Research Institute, Créteil, France
| | - Olivier Schwartz
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, Paris, France
- Vaccine Research Institute, Créteil, France
| | - Ann J. Hessell
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Margaret E. Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, New Hampshire, USA
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
15
|
Rodríguez-Ramírez KT, Norte-Muñoz M, Lucas-Ruiz F, Gallego-Ortega A, Calzaferri F, García-Bernal D, Martínez CM, Galindo-Romero C, de los Ríos C, Vidal-Sanz M, Agudo-Barriuso M. Retinal response to systemic inflammation differs between sexes and neurons. Front Immunol 2024; 15:1340013. [PMID: 38384465 PMCID: PMC10880026 DOI: 10.3389/fimmu.2024.1340013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/23/2024] [Indexed: 02/23/2024] Open
Abstract
Background Neurological dysfunction and glial activation are common in severe infections such as sepsis. There is a sexual dimorphism in the response to systemic inflammation in both patients and animal models, but there are few comparative studies. Here, we investigate the effect of systemic inflammation induced by intraperitoneal administration of lipopolysaccharide (LPS) on the retina of male and female mice and determine whether antagonism of the NLRP3 inflammasome and the extrinsic pathway of apoptosis have protective effects on the retina. Methods A single intraperitoneal injection of LPS (5 mg/kg) was administered to two months old C57BL/6J male and female mice. Retinas were examined longitudinally in vivo using electroretinography and spectral domain optical coherence tomography. Retinal ganglion cell (RGC) survival and microglial activation were analysed in flat-mounts. Retinal extracts were used for flow cytometric analysis of CD45 and CD11b positive cells. Matched plasma and retinal levels of proinflammatory cytokines were measured by ELISA. Retinal function and RGC survival were assessed in animals treated with P2X7R and TNFR1 antagonists alone or in combination. Results In LPS-treated animals of both sexes, there was transient retinal dysfunction, loss of vision-forming but not non-vision forming RGCs, retinal swelling, microglial activation, cell infiltration, and increases in TNF and IL-1β. Compared to females, males showed higher vision-forming RGC death, slower functional recovery, and overexpression of lymphotoxin alpha in their retinas. P2X7R and TNFR1 antagonism, alone or in combination, rescued vision-forming RGCs. P2X7R antagonism also rescued retinal function. Response to treatment was better in females than in males. Conclusions Systemic LPS has neuronal and sex-specific adverse effects in the mouse retina, which are counteracted by targeting the NLRP3 inflammasome and the extrinsic pathway of apoptosis. Our results highlight the need to analyse males and females in preclinical studies of inflammatory diseases affecting the central nervous system.
Collapse
Affiliation(s)
- Kristy T. Rodríguez-Ramírez
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - María Norte-Muñoz
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Fernando Lucas-Ruiz
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Alejandro Gallego-Ortega
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Francesco Calzaferri
- Instituto-Fundación Teófilo Hernando and Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - David García-Bernal
- Grupo de Trasplante Hematopoyético y Terapia Celular, Departamento de Bioquímica y Biología Molecular B e Inmunología, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Carlos M. Martínez
- Plataforma de Patología, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Caridad Galindo-Romero
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Cristóbal de los Ríos
- Instituto-Fundación Teófilo Hernando and Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - Manuel Vidal-Sanz
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Marta Agudo-Barriuso
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| |
Collapse
|
16
|
Wen L, Bi D, Shen Y. Complement-mediated synapse loss in Alzheimer's disease: mechanisms and involvement of risk factors. Trends Neurosci 2024; 47:135-149. [PMID: 38129195 DOI: 10.1016/j.tins.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
The complement system is increasingly recognized as a key player in the synapse loss and cognitive impairments observed in Alzheimer's disease (AD). In particular, the process of complement-dependent synaptic pruning through phagocytosis is over-activated in AD brains, driving detrimental excessive synapse elimination and contributing to synapse loss, which is the strongest neurobiological correlate of cognitive impairments in AD. Herein we review recent advances in characterizing complement-mediated synapse loss in AD, summarize the underlying mechanisms, and discuss the possible involvement of AD risk factors such as aging and various risk genes. We conclude with an overview of key questions that remain to be addressed.
Collapse
Affiliation(s)
- Lang Wen
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Danlei Bi
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, 230026, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230026, China; CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230026, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Yong Shen
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, 230026, China; CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230026, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
17
|
Ricardo-da-Silva FY, Armstrong-Jr R, Ramos MMDA, Vidal-Dos-Santos M, Jesus Correia C, Ottens PJ, Moreira LFP, Leuvenink HGD, Breithaupt-Faloppa AC. Male versus female inflammatory response after brain death model followed by ex vivo lung perfusion. Biol Sex Differ 2024; 15:11. [PMID: 38287395 PMCID: PMC10826050 DOI: 10.1186/s13293-024-00581-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 01/02/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Ex vivo lung perfusion (EVLP) is a useful tool for assessing lung grafts quality before transplantation. Studies indicate that donor sex is as an important factor for transplant outcome, as females present higher inflammatory response to brain death (BD) than males. Here, we investigated sex differences in the lungs of rats subjected to BD followed by EVLP. METHODS Male and female Wistar rats were subjected to BD, and as controls sham animals. Arterial blood was sampled for gas analysis. Heart-lung blocks were kept in cold storage (1 h) and normothermic EVLP carried out (4 h), meanwhile ventilation parameters were recorded. Perfusate was sampled for gas analysis and IL-1β levels. Leukocyte infiltration, myeloperoxidase presence, IL-1β gene expression, and long-term release in lung culture (explant) were evaluated. RESULTS Brain dead females presented a low lung function after BD, compared to BD-males; however, at the end of the EVLP period oxygenation capacity decreased in all BD groups. Overall, ventilation parameters were maintained in all groups. After EVLP lung infiltrate was higher in brain dead females, with higher neutrophil content, and accompanied by high IL-1β levels, with increased gene expression and concentration in the culture medium (explant) 24 h after EVLP. Female rats presented higher lung inflammation after BD than male rats. Despite maintaining lung function and ventilation mechanics parameters for 4 h, EVLP was not able to alter this profile. CONCLUSION In this context, further studies should focus on therapeutic measures to control inflammation in donor or during EVLP to increase lung quality.
Collapse
Affiliation(s)
- Fernanda Yamamoto Ricardo-da-Silva
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), HC-FMUSP, Instituto Do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 2º Andar, Sala 2146, São Paulo, 01246-903, Brazil
| | - Roberto Armstrong-Jr
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), HC-FMUSP, Instituto Do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 2º Andar, Sala 2146, São Paulo, 01246-903, Brazil
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Mayara Munhoz de Assis Ramos
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), HC-FMUSP, Instituto Do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 2º Andar, Sala 2146, São Paulo, 01246-903, Brazil
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Marina Vidal-Dos-Santos
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), HC-FMUSP, Instituto Do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 2º Andar, Sala 2146, São Paulo, 01246-903, Brazil
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Cristiano Jesus Correia
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), HC-FMUSP, Instituto Do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 2º Andar, Sala 2146, São Paulo, 01246-903, Brazil
| | - Petra J Ottens
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Luiz Felipe Pinho Moreira
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), HC-FMUSP, Instituto Do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 2º Andar, Sala 2146, São Paulo, 01246-903, Brazil
| | - Henri G D Leuvenink
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Ana Cristina Breithaupt-Faloppa
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), HC-FMUSP, Instituto Do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 2º Andar, Sala 2146, São Paulo, 01246-903, Brazil.
| |
Collapse
|
18
|
Hosseinzadeh S, Afshari S, Molaei S, Rezaei N, Dadkhah M. The role of genetics and gender specific differences in neurodegenerative disorders: Insights from molecular and immune landscape. J Neuroimmunol 2023; 384:578206. [PMID: 37813041 DOI: 10.1016/j.jneuroim.2023.578206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/09/2023] [Accepted: 09/23/2023] [Indexed: 10/11/2023]
Abstract
Neurodegenerative disorders (NDDs) are the most common neurological disorders with high prevalence and have significant socioeconomic implications. Understanding the underlying cellular and molecular mechanisms associated with the immune system can be effective in disease etiology, leading to more effective therapeutic approaches for both females and males. The central nervous system (CNS) actively participates in immune responses, both within and outside the CNS. Immune system activation is a common feature in NDDs. Gender-specific factors play a significant role in the prevalence, progression, and manifestation of NDDs. Neuroinflammation, in both inflammatory neurological and neurodegenerative conditions, is defined by the triggering of microglia and astrocyte cell activation. This results in the secretion of pro-inflammatory cytokines and chemokines. Numerous studies have documented the role of neuroinflammation in neurological diseases, highlighting the involvement of immune signaling pathways in disease development. Converging evidence support immune system involvement during neurodegeneration in NDDs. In this review, we summarize emerging evidence that reveals gender-dependent differences in immune responses related to NDDs. Also, we highlight sex differences in immune responses and discuss how these sex-specific influences can increase the risk of NDDs. Understanding the role of gender-specific factors can aid in developing targeted therapeutic strategies and improving patient outcomes. Ultimately, the better understanding of these mechanisms contributed to sex-dependent immune response in NDDs, can be critically usful in targeting of immune signaling cascades in such disorders. In this regard, sex-related immune responses in NDDs may be promising and effective targets in therapeutic strategies.
Collapse
Affiliation(s)
- Shahnaz Hosseinzadeh
- Department of Microbiology & Immunology, School of Medicine, Ardabil University of Medical Sciences, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Salva Afshari
- Students Research Committee, Pharmacy School, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Soheila Molaei
- Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran 1419733151, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education Research Network (USERN), Tehran, Iran
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
19
|
Kelkar NS, Goldberg BS, Dufloo J, Bruel T, Schwartz O, Hessell AJ, Ackerman ME. Sex and species associated differences in Complement-mediated immunity in Humans and Rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563614. [PMID: 37961263 PMCID: PMC10634758 DOI: 10.1101/2023.10.23.563614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The complement system can be viewed as a 'moderator' of innate immunity, 'instructor' of humoral immunity, and 'regulator' of adaptive immunity. While sex and aging are known to affect humoral and cellular immune systems, their impact on the complement pathway in humans and rhesus macaques, a commonly used non-human primate model system, have not been well-studied. To address this knowledge gap, we analyzed serum samples from 90 humans and 75 rhesus macaques for the abundance and activity of the complement system components. While sequences of cascade proteins were highly conserved, dramatically different levels were observed between species. Whereas the low levels detected in rhesus samples raised questions about the suitability of the test, differences in levels of complement proteins were observed in male and female humans. Levels of total and antibody-dependent deposition of C1q and C3b on a glycosylated antigen differed between human and rhesus, suggesting differential recognition of glycans. Functional differences in complement-mediated lysis of antibody-sensitized cells were observed in multiple assays and showed that human females frequently exhibited higher lytic activity than human males or rhesus macaques, which typically did not exhibit such sexual dimorphism. Other differences between species and sexes were observed in more narrow contexts-for only certain antibodies, antigens, or assays. Collectively, these results expand our knowledge of sexual dimorphism in the complement system in humans, identifying differences that appear to be absent from rhesus macaques.
Collapse
Affiliation(s)
- Natasha S. Kelkar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Benjamin S. Goldberg
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
- Present Address: Metaphore Biotechnologies Inc., Cambridge, MA, USA
| | - Jérémy Dufloo
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, 75015 Paris, France
- Present Address: Institute for Integrative Systems Biology (I2SysBio), Universitat da Valencia-CSIC, 46980 Valencia, Spain
| | - Timothée Bruel
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, 75015 Paris, France
- Vaccine Research Institute, 9400 Créteil, France
| | - Olivier Schwartz
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus and Immunity Unit, 75015 Paris, France
- Vaccine Research Institute, 9400 Créteil, France
| | - Ann J. Hessell
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Margaret E. Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| |
Collapse
|
20
|
Wen L, Yang X, Wu Z, Fu S, Zhan Y, Chen Z, Bi D, Shen Y. The complement inhibitor CD59 is required for GABAergic synaptic transmission in the dentate gyrus. Cell Rep 2023; 42:112349. [PMID: 37027303 DOI: 10.1016/j.celrep.2023.112349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 01/31/2023] [Accepted: 03/21/2023] [Indexed: 04/08/2023] Open
Abstract
Complement-dependent microglia pruning of excitatory synapses has been widely reported in physiological and pathological conditions, with few reports concerning pruning of inhibitory synapses or direct regulation of synaptic transmission by complement components. Here, we report that loss of CD59, an important endogenous inhibitor of the complement system, leads to compromised spatial memory performance. Furthermore, CD59 deficiency impairs GABAergic synaptic transmission in the hippocampal dentate gyrus (DG). This depends on regulation of GABA release triggered by Ca2+ influx through voltage-gated calcium channels (VGCCs) rather than inhibitory synaptic pruning by microglia. Notably, CD59 colocalizes with inhibitory pre-synaptic terminals and regulates SNARE complex assembly. Together, these results demonstrate that the complement regulator CD59 plays an important role in normal hippocampal function.
Collapse
Affiliation(s)
- Lang Wen
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Xiaoli Yang
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Zujun Wu
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Shumei Fu
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yaxi Zhan
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Zuolong Chen
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215000, China
| | - Danlei Bi
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei 230026, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230026, China; CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yong Shen
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
21
|
Olivieri F, Marchegiani F, Matacchione G, Giuliani A, Ramini D, Fazioli F, Sabbatinelli J, Bonafè M. Sex/gender-related differences in inflammaging. Mech Ageing Dev 2023; 211:111792. [PMID: 36806605 DOI: 10.1016/j.mad.2023.111792] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023]
Abstract
Geroscience puts mechanisms of aging as a driver of the most common age-related diseases and dysfunctions. Under this perspective, addressing the basic mechanisms of aging will produce a better understanding than addressing each disease pathophysiology individually. Worldwide, despite greater functional impairment, life expectancy is higher in women than in men. Gender differences in the prevalence of multimorbidity lead mandatory to the understanding of the mechanisms underlying gender-related differences in multimorbidity patterns and disability-free life expectancy. Extensive literature suggested that inflammaging is at the crossroad of aging and age-related diseases. In this review, we highlight the main evidence on sex/gender differences in the mechanisms that foster inflammaging, i.e. the age-dependent triggering of innate immunity, modifications of adaptive immunity, and accrual of senescent cells, underpinning some biomarkers of inflammaging that show sex-related differences. In the framework of the "gender medicine perspective", we will also discuss how sex/gender differences in inflammaging can affect sex differences in COVID-19 severe outcomes.
Collapse
Affiliation(s)
- Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
| | | | - Giulia Matacchione
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Angelica Giuliani
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Deborah Ramini
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
| | - Francesca Fazioli
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Laboratory Medicine Unit, Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy.
| | - Massimiliano Bonafè
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| |
Collapse
|
22
|
de Boer ECW, Thielen AJF, Langereis JD, Kamp A, Brouwer MC, Oskam N, Jongsma ML, Baral AJ, Spaapen RM, Zeerleder S, Vidarsson G, Rispens T, Wouters D, Pouw RB, Jongerius I. The contribution of the alternative pathway in complement activation on cell surfaces depends on the strength of classical pathway initiation. Clin Transl Immunology 2023; 12:e1436. [PMID: 36721662 PMCID: PMC9881211 DOI: 10.1002/cti2.1436] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 10/31/2022] [Accepted: 12/23/2022] [Indexed: 01/28/2023] Open
Abstract
Objectives The complement system is an important component of innate immunity. The alternative pathway (AP) amplification loop is considered an essential feed forward mechanism for complement activation. However, the role of the AP in classical pathway (CP) activation has only been studied in ELISA settings. Here, we investigated its contribution on physiologically relevant surfaces of human cells and bacterial pathogens and in antibody-mediated complement activation, including in autoimmune haemolytic anaemia (AIHA) setting with autoantibodies against red blood cells (RBCs). Methods We evaluated the contribution of the AP to complement responses initiated through the CP on human RBCs by serum of AIHA patients and recombinant antibodies. Moreover, we studied complement activation on Neisseria meningitidis and Escherichia coli. The effect of the AP was examined using either AP-depleted sera or antibodies against factor B and factor D. Results We show that the amplification loop is redundant when efficient CP activation takes place. This is independent of the presence of membrane-bound complement regulators. The role of the AP may become significant when insufficient CP complement activation occurs, but this depends on antibody levels and (sub)class. Our data indicate that therapeutic intervention in the amplification loop will most likely not be effective to treat antibody-mediated diseases. Conclusion The AP can be bypassed through efficient CP activation. The AP amplification loop has a role in complement activation during conditions of modest activation via the CP, when it can allow for efficient complement-mediated killing.
Collapse
Affiliation(s)
- Esther CW de Boer
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Department of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's HospitalAmsterdam University Medical CentreAmsterdamThe Netherlands
| | - Astrid JF Thielen
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Jeroen D Langereis
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life SciencesRadboudumcNijmegenThe Netherlands,Radboud Center for Infectious Diseases, RadboudumcNijmegenThe Netherlands
| | - Angela Kamp
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Mieke C Brouwer
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Nienke Oskam
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Marlieke L Jongsma
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - April J Baral
- Translational and Clinical Research InstituteNewcastle upon TyneUK
| | - Robbert M Spaapen
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Sacha Zeerleder
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Department of Hematology, Luzerner KantonsspitalLuzern and University of BernBernSwitzerland,Department for BioMedical ResearchUniversity of BernBernSwitzerland
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner LaboratoryAmsterdam University Medical CenterAmsterdamThe Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands
| | - Diana Wouters
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Centre for Infectious Disease ControlNational Institute for Public Health and the Environment (RIVM)BilthovenThe Netherlands
| | - Richard B Pouw
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Sanquin Health SolutionsAmsterdamThe Netherlands
| | - Ilse Jongerius
- Department of Immunopathology, Sanquin Research and Landsteiner LaboratoryAmsterdam Infection and Immunity Institute, Amsterdam University Medical CentreAmsterdamThe Netherlands,Department of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's HospitalAmsterdam University Medical CentreAmsterdamThe Netherlands
| |
Collapse
|
23
|
Functional Loss of Terminal Complement Complex Protects Rabbits from Injury-Induced Osteoarthritis on Structural and Cellular Level. Biomolecules 2023; 13:biom13020216. [PMID: 36830586 PMCID: PMC9953363 DOI: 10.3390/biom13020216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023] Open
Abstract
The terminal complement complex (TCC) has been described as a potential driver in the pathogenesis of posttraumatic osteoarthritis (PTOA). However, sublytic TCC deposition might also play a crucial role in bone development and regeneration. Therefore, we elucidated the effects of TCC on joint-related tissues using a rabbit PTOA model. In brief, a C6-deficient rabbit breed was characterized on genetic, protein, and functional levels. Anterior cruciate ligament transection (ACLT) was performed in C6-deficient (C6-/-) and C6-sufficient (C6+/-) rabbits. After eight weeks, the progression of PTOA was determined histologically. Moreover, the structure of the subchondral bone was evaluated by µCT analysis. C6 deficiency could be attributed to a homozygous 3.6 kb deletion within the C6 gene and subsequent loss of the C5b binding site. Serum from C6-/- animals revealed no hemolytic activity. After ACLT surgery, joints of C6-/- rabbits exhibited significantly lower OA scores, including reduced cartilage damage, hypocellularity, cluster formation, and osteophyte number, as well as lower chondrocyte apoptosis rates and synovial prostaglandin E2 levels. Moreover, ACLT surgery significantly decreased the trabecular number in the subchondral bone of C6-/- rabbits. Overall, the absence of TCC protected from injury-induced OA progression but had minor effects on the micro-structure of the subchondral bone.
Collapse
|
24
|
Dodd KC, Menon M. Sex bias in lymphocytes: Implications for autoimmune diseases. Front Immunol 2022; 13:945762. [PMID: 36505451 PMCID: PMC9730535 DOI: 10.3389/fimmu.2022.945762] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
Autoimmune diseases are characterized by a significant sex dimorphism, with women showing increased susceptibility to disease. This is, at least in part, due to sex-dependent differences in the immune system that are influenced by the complex interplay between sex hormones and sex chromosomes, with contribution from sociological factors, diet and gut microbiota. Sex differences are evident in the number and function of lymphocyte populations. Women mount a stronger pro-inflammatory response than males, with increased lymphocyte proliferation, activation and pro-inflammatory cytokine production, whereas men display expanded regulatory cell subsets. Ageing alters the immune landscape of men and women in differing ways, resulting in changes in autoimmune disease susceptibility. Here we review the current literature on sex differences in lymphocyte function, the factors that influence this, and the implications for autoimmune disease. We propose that improved understanding of sex bias in lymphocyte function can provide sex-specific tailoring of treatment strategies for better management of autoimmune diseases.
Collapse
Affiliation(s)
- Katherine C. Dodd
- Lydia Becker Institute of Immunology and Inflammation, Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom,Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Salford, United Kingdom
| | - Madhvi Menon
- Lydia Becker Institute of Immunology and Inflammation, Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom,*Correspondence: Madhvi Menon,
| |
Collapse
|
25
|
Crowley MA, Garland DL, Sellner H, Banks A, Fan L, Rejtar T, Buchanan N, Delgado O, Xu YY, Jose S, Adams CM, Mogi M, Wang K, Bigelow CE, Poor S, Anderson K, Jaffee BD, Prasanna G, Grosskreutz C, Fernandez-Godino R, Pierce EA, Dryja TP, Liao SM. Complement factor B is critical for sub-RPE deposit accumulation in a model of Doyne honeycomb retinal dystrophy with features of age-related macular degeneration. Hum Mol Genet 2022; 32:204-217. [PMID: 35943778 PMCID: PMC9840207 DOI: 10.1093/hmg/ddac187] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/04/2022] [Accepted: 08/05/2022] [Indexed: 01/19/2023] Open
Abstract
EFEMP1 R345W is a dominant mutation causing Doyne honeycomb retinal dystrophy/malattia leventinese (DHRD/ML), a rare blinding disease with clinical pathology similar to age-related macular degeneration (AMD). Aged Efemp1 R345W/R345W knock-in mice (Efemp1ki/ki) develop microscopic deposits on the basal side of retinal pigment epithelial cells (RPE), an early feature in DHRD/ML and AMD. Here, we assessed the role of alternative complement pathway component factor B (FB) in the formation of these deposits. RNA-seq analysis of the posterior eyecups revealed increased unfolded protein response, decreased mitochondrial function in the neural retina (by 3 months of age) and increased inflammatory pathways in both neural retina and posterior eyecups (at 17 months of age) of Efemp1ki/ki mice compared with wild-type littermate controls. Proteomics analysis of eye lysates confirmed similar dysregulated pathways as detected by RNA-seq. Complement activation was increased in aged Efemp1ki/ki eyes with an approximately 2-fold elevation of complement breakdown products iC3b and Ba (P < 0.05). Deletion of the Cfb gene in female Efemp1ki/ki mice partially normalized the above dysregulated biological pathway changes and oral dosing of a small molecule FB inhibitor from 10 to 12 months of age reduced sub-RPE deposits by 65% (P = 0.029). In contrast, male Efemp1ki/ki mice had fewer sub-RPE deposits than age-matched females, no elevation of ocular complement activation and no effect of FB inhibition on sub-RPE deposits. The effects of FB deletion or inhibition on Efemp1ki/ki mice supports systemic inhibition of the alternative complement pathway as a potential treatment of dry AMD and DHRD/ML.
Collapse
Affiliation(s)
- Maura A Crowley
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Donita L Garland
- Ocular Genomics Institute at Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Holger Sellner
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Angela Banks
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Lin Fan
- Analytical Sciences and Imaging, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Tomas Rejtar
- Analytical Sciences and Imaging, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Natasha Buchanan
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Omar Delgado
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Yong Yao Xu
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Sandra Jose
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Christopher M Adams
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Muneto Mogi
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Karen Wang
- Analytical Sciences and Imaging, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Chad E Bigelow
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Stephen Poor
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | | | - Bruce D Jaffee
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Ganesh Prasanna
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Cynthia Grosskreutz
- Ophthalmology, Novartis Institutes for BioMedical Research, Cambridge, MA 02319, USA
| | - Rosario Fernandez-Godino
- Ocular Genomics Institute at Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Eric A Pierce
- Ocular Genomics Institute at Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | | | - Sha-Mei Liao
- To whom correspondence should be addressed at: Department of Ophthalmology, Novartis Institutes for BioMedical Research, 22 Windsor Street, Cambridge, MA 02139, USA. Tel: +1-(617)871-4004; Fax: +1-(617)871-5748;
| |
Collapse
|
26
|
Sodhi EU, Philpott HT, Carter MM, Birmingham TB, Appleton CT. Sex-Differences and Associations Between Complement Activation and Synovial Vascularization in Patients with Late-Stage Knee Osteoarthritis. Front Immunol 2022; 13:890094. [PMID: 35686134 PMCID: PMC9170895 DOI: 10.3389/fimmu.2022.890094] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 04/27/2022] [Indexed: 11/17/2022] Open
Abstract
Purpose Synovial inflammation in knee osteoarthritis (OA) causes disorganized synovial angiogenesis and complement activation in synovial fluid, but links between complement and synovial microvascular pathology have not been established. Since complement causes vascular pathology in other diseases and since sex-differences exist in complement activation and in OA, we investigated sex differences in synovial fluid complement factors, synovial tissue vascular pathology, and associations between complement and synovial vascular pathology in patients with late-stage knee OA. Methods Patients with symptomatic, late-stage radiographic knee OA undergoing total knee arthroplasty or high tibial osteotomy provided matched synovial fluid and tissue biopsies during surgery. Complement factors (C2, C5, adipsin, MBL, and CFI) and terminal complement complex (sC5b-C9) were measured in synovial fluid by multiplex or enzyme-linked immunosorbent assay, respectively. Features of synovial vascular pathology (vascularization, perivascular edema, and vasculopathy) were assessed by histopathology. Multivariate linear regression models were used to assess associations between synovial fluid complement factors and histopathological features of vascular pathology, with adjustment for age, sex, body mass index, and sex interaction. Sex-disaggregated comparisons were completed. Results Synovial fluid biomarker and histopathology data were included from 97 patients. Most synovial fluid complement factors and synovial tissue histopathological features were similar between sexes. Synovial fluid C5 trended to lower levels in males (-20.93 ng/mL [95%CI -42.08, 0.23] p=0.05). Median vasculopathy scores (0.42 [95%CI 0.07, 0.77] p=0.02) were higher in males. In the full cohort, C5 concentration was associated with lower vascularization scores (-0.005 [95%CI -0.010, -0.0001] p=0.04) while accounting for sex*C5 interaction. In sex-disaggregated analyses, increased C5 concentration was associated with lower vascularization scores (-0.005 [95%CI –0.009, -0.0001] p=0.04) in male patients, but not in female patients. Males had higher sC5b-C9 compared to females. Additionally, males with high C5 had a higher synovial fluid concentration of sC5b-C9 compared to males with low C5. No differences were found in females. Conclusion Higher synovial fluid C5 levels were associated with increased complement activation and decreased synovial vascularization in males but not in females with OA. Future studies should test whether synovial fluid complement activation suppresses synovial angiogenesis and identify mechanisms accounting for C5-related sex-differences in synovial fluid complement activation in patients with knee OA.
Collapse
Affiliation(s)
- Emily U Sodhi
- Department of Physiology & Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Holly T Philpott
- Health & Rehabilitation Sciences, Faculty of Health Sciences, Western University, London, ON, Canada.,Bone & Joint Institute, Western University, London, ON, Canada
| | - McKenzie M Carter
- Department of Physiology & Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Bone & Joint Institute, Western University, London, ON, Canada
| | - Trevor B Birmingham
- Health & Rehabilitation Sciences, Faculty of Health Sciences, Western University, London, ON, Canada.,Bone & Joint Institute, Western University, London, ON, Canada
| | - C Thomas Appleton
- Department of Physiology & Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Health & Rehabilitation Sciences, Faculty of Health Sciences, Western University, London, ON, Canada.,Bone & Joint Institute, Western University, London, ON, Canada.,Department of Medicine, Schulich School of Medicine, Western University, London, ON, Canada
| |
Collapse
|
27
|
Westacott LJ, Wilkinson LS. Complement Dependent Synaptic Reorganisation During Critical Periods of Brain Development and Risk for Psychiatric Disorder. Front Neurosci 2022; 16:840266. [PMID: 35600620 PMCID: PMC9120629 DOI: 10.3389/fnins.2022.840266] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Abstract
We now know that the immune system plays a major role in the complex processes underlying brain development throughout the lifespan, carrying out a number of important homeostatic functions under physiological conditions in the absence of pathological inflammation or infection. In particular, complement-mediated synaptic pruning during critical periods of early life may play a key role in shaping brain development and subsequent risk for psychopathology, including neurodevelopmental disorders such as schizophrenia and autism spectrum disorders. However, these disorders vary greatly in their onset, disease course, and prevalence amongst sexes suggesting complex interactions between the immune system, sex and the unique developmental trajectories of circuitries underlying different brain functions which are yet to be fully understood. Perturbations of homeostatic neuroimmune interactions during different critical periods in which regional circuits mature may have a plethora of long-term consequences for psychiatric phenotypes, but at present there is a gap in our understanding of how these mechanisms may impact on the structural and functional changes occurring in the brain at different developmental stages. In this article we will consider the latest developments in the field of complement mediated synaptic pruning where our understanding is beginning to move beyond the visual system where this process was first described, to brain areas and developmental periods of potential relevance to psychiatric disorders.
Collapse
Affiliation(s)
- Laura J. Westacott
- Neuroscience and Mental Health Innovation Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Lawrence S. Wilkinson
- Neuroscience and Mental Health Innovation Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Behavioural Genetics Group, Schools of Psychology and Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
28
|
Marin AI, Poppelaars F, Wagner BD, Palestine AG, Patnaik JL, Holers VM, Frazer-Abel AA, Mathias MT, Manoharan N, Fonteh CN, Mandava N, Lynch AM. Sex and Age-Related Differences in Complement Factors Among Patients With Intermediate Age-Related Macular Degeneration. Transl Vis Sci Technol 2022; 11:22. [PMID: 35594041 PMCID: PMC9145081 DOI: 10.1167/tvst.11.5.22] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Purpose Age-related macular degeneration (AMD) is an acquired degenerative disease of the retina classified into early, intermediate, and advanced AMD. A key factor in the pathogenesis of AMD is the complement system. The interaction of age and sex with the complement system may affect the risk of developing AMD. The purpose of this study was to determine if there were sex-specific differences in levels of complement factors among patients with the intermediate phenotype of AMD (iAMD) and explore the correlation between age and complement proteins. Methods We studied complement factors in patients with iAMD and controls without AMD. Nonparametric, rank-based linear regressions including a sex by AMD interaction were used to compare levels for each analyte. Correlations between age and complement proteins were evaluated using the Spearman rank correlation coefficient. Results We found significantly higher levels of factor B and factor I in females compared with males with iAMD, whereas no differences were seen in complement levels in male and female controls. The ratios of Ba/factor B, C3a/C3, C4b/C4, and C5a/C5 were not different in males and females with iAMD. Conclusions We demonstrate disparities in a subset of systemic complement factors between females and males with iAMD, but apparent complement turnover as measured by ratios of activation fragments to intact molecules was not different between these groups. The results suggest that complement system levels, including complement regulator factor I, exhibits sex-related differences in patients with iAMD and highlights that stratification by sex might be helpful in the interpretation of clinical trials of anticomplement therapy.
Collapse
Affiliation(s)
- A Itzam Marin
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Felix Poppelaars
- Division of Nephrology and Hypertension, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Brandie D Wagner
- Department of Biostatistics and Informatics, University of Colorado School of Public Health, Aurora, Colorado, USA
| | - Alan G Palestine
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Jennifer L Patnaik
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - V Michael Holers
- Departments of Medicine and Immunology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Ashley A Frazer-Abel
- Exsera BioLabs, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Marc T Mathias
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Niranjan Manoharan
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Cheryl N Fonteh
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Naresh Mandava
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Anne M Lynch
- Department of Ophthalmology, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | | |
Collapse
|
29
|
Panaampon J, Kariya R, Okada S. Efficacy and mechanism of the anti-CD38 monoclonal antibody Daratumumab against primary effusion lymphoma. Cancer Immunol Immunother 2022; 71:1017-1031. [PMID: 34545416 PMCID: PMC10991639 DOI: 10.1007/s00262-021-03054-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 09/06/2021] [Indexed: 02/08/2023]
Abstract
Primary effusion lymphoma (PEL) is a rare, aggressive B cell non-Hodgkin's lymphoma of the body cavities with malignant effusions. The prognosis is poor, and no optimal treatment has been established. CD38 is a type II transmembrane glycoprotein known to overexpress in multiple myeloma (MM). Daratumumab (DARA), a human CD38-targeting monoclonal antibody (mAb), is approved for MM treatment. In this study, we found expression of CD38 on PEL cells and assessed the anti-PEL activity of DARA. We found that both KHYG-1 and N6 (CD16-transfected KHYG-1) NK cell lines showed direct killing activity against PEL cells with induction of CD107a, and NK-mediated cytotoxicity by N6NK (CD16+) cells increased with DARA treatment. We confirmed direct NK activity and antibody-dependent cell cytotoxicity (ADCC) by expanded NK cells, indicating that DARA has high ADCC activity. We elucidated the antibody-dependent cell phagocytosis (ADCP) by using human monocyte-derived macrophages (MDMs) and mouse peritoneal macrophages. DARA also showed potent complement-dependent cytolysis (CDC) toward PEL. DARA also induced PEL cell death in the presence of a cross-linking antibody. Moreover, treatment with DARA inhibited tumor growth in a PEL xenograft mouse model. These results provide preclinical evidence that Ab targeting of CD38 could be an effective therapeutic strategy for the treatment of PEL.
Collapse
Affiliation(s)
- Jutatip Panaampon
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Ryusho Kariya
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan.
| |
Collapse
|
30
|
Zanchi C, Locatelli M, Cerullo D, Aumiller V, Corna D, Rottoli D, Eisermann M, Donadelli R, Mousavi M, Noris M, Remuzzi G, Benigni A, Zoja C. Therapeutic Small Interfering RNA Targeting Complement C3 in a Mouse Model of C3 Glomerulopathy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1772-1781. [PMID: 35277417 DOI: 10.4049/jimmunol.2100730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 01/28/2022] [Indexed: 06/14/2023]
Abstract
Alternative pathway complement dysregulation with abnormal glomerular C3 deposits and glomerular damage is a key mechanism of pathology in C3 glomerulopathy (C3G). No disease-specific treatments are currently available for C3G. Therapeutics inhibiting complement are emerging as a potential strategy for the treatment of C3G. In this study, we investigated the effects of N-acetylgalactosamine (GalNAc)-conjugated small interfering RNA (siRNA) targeting the C3 component of complement that inhibits liver C3 expression in the C3G model of mice with heterozygous deficiency of factor H (Cfh +/- mice). We showed a duration of action for GalNAc-conjugated C3 siRNA in reducing the liver C3 gene expression in Cfh +/- mice that were dosed s.c. once a month for up to 7 mo. C3 siRNA limited fluid-phase alternative pathway activation, reducing circulating C3 fragmentation and activation of factor B. Treatment with GalNAc-conjugated C3 siRNA reduced glomerular C3d deposits in Cfh +/- mice to levels similar to those of wild-type mice. Ultrastructural analysis further revealed the efficacy of the C3 siRNA in slowing the formation of mesangial and subendothelial electron-dense deposits. The present data indicate that RNA interference-mediated C3 silencing in the liver may be a relevant therapeutic strategy for treating patients with C3G associated with the haploinsufficiency of complement factor H.
Collapse
Affiliation(s)
- Cristina Zanchi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy; and
| | - Monica Locatelli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy; and
| | - Domenico Cerullo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy; and
| | | | - Daniela Corna
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy; and
| | - Daniela Rottoli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy; and
| | | | - Roberta Donadelli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy; and
| | - Mansoureh Mousavi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy; and
| | - Marina Noris
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy; and
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy; and
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy; and
| | - Carlamaria Zoja
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy; and
| |
Collapse
|
31
|
Dubois E, Galindo AN, Dayon L, Cominetti O. Assessing normalization methods in mass spectrometry-based proteome profiling of clinical samples. Biosystems 2022; 215-216:104661. [PMID: 35247480 DOI: 10.1016/j.biosystems.2022.104661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 02/21/2022] [Accepted: 02/28/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Large-scale proteomic studies have to deal with unwanted variability, especially when samples originate from different centers and multiple analytical batches are needed. Such variability is typically added throughout all the steps of a clinical research study, from human biological sample collection and storage, sample preparation, spectral data acquisition, to peptide and protein quantification. In order to remove such diverse and unwanted variability, normalization of the protein data is performed. There have been already several published reviews comparing normalization methods in the -omics field, but reports focusing on proteomic data generated with mass spectrometry (MS) are much fewer. Additionally, most of these reports have only dealt with small datasets. RESULTS As a case study, here we focused on the normalization of a large MS-based proteomic dataset obtained from an overweight and obese pan-European cohort, where different normalization methods were evaluated, namely: center standardize, quantile protein, quantile sample, global standardization, ComBat, median centering, mean centering, single standard and removal of unwanted variation (RUV); some of these are generic normalization methods while others have been specifically created to deal with genomic or metabolomic data. We checked how relationships between proteins and clinical variables (e.g., gender, levels of triglycerides or cholesterol) were improved after normalizing the data with the different methods. CONCLUSIONS Some normalization methods were better adapted for this particular large-scale shotgun proteomic dataset of human plasma samples labeled with isobaric tags and analyzed with liquid chromatography-tandem MS. In particular, quantile sample normalization, RUV, mean and median centering showed very good performance, while quantile protein normalization provided worse results than those obtained with unnormalized data.
Collapse
Affiliation(s)
- Etienne Dubois
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, EPFL Innovation Park, 1015, Lausanne, Switzerland
| | - Antonio Núñez Galindo
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, EPFL Innovation Park, 1015, Lausanne, Switzerland
| | - Loïc Dayon
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, EPFL Innovation Park, 1015, Lausanne, Switzerland; Chemistry and Chemical Engineering Section, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Ornella Cominetti
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, EPFL Innovation Park, 1015, Lausanne, Switzerland.
| |
Collapse
|
32
|
Circulating C1q levels in health and disease, more than just a biomarker. Mol Immunol 2021; 140:206-216. [PMID: 34735869 DOI: 10.1016/j.molimm.2021.10.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/03/2021] [Accepted: 10/11/2021] [Indexed: 12/21/2022]
Abstract
C1q is the recognition molecule of the classical pathway of the complement system. By binding to its targets, such as antigen-bound immunoglobulins or C-reactive protein, C1q contributes to the innate defense against infections. However, C1q also plays several other roles beyond its traditional role in complement activation. Circulating levels of C1q are determined in routine diagnostics as biomarker in several diseases. Decreased C1q levels are present in several autoimmune conditions. The decreased levels reflect the consumption of C1q by complement activation and serves as a biomarker for disease activity. In contrast, increased C1q levels are present in infectious and inflammatory diseases and may serve as a diagnostic biomarker. The increased levels of C1q are still incompletely understood but are suggested to modulate the adaptive immune response as C1q is known to impact on the maturation status of antigen-presenting cells and C1q impacts directly on T cells leading to decreased T-cell activity in high C1q conditions. In this review, we provide a comprehensive overview of the current literature on circulating levels of C1q in health and disease, and discuss how C1q can both protect against infections as well as maintain tolerance by regulating adaptive immunity.
Collapse
|
33
|
Zelek WM. Measuring Total Classical Pathway and Activities of Individual Components of the Mouse Complement Pathway. Bio Protoc 2021; 11:e4175. [PMID: 34722822 DOI: 10.21769/bioprotoc.4175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/04/2021] [Accepted: 07/09/2021] [Indexed: 01/13/2023] Open
Abstract
The complement system is a central component of innate immunity, responsible for recognition and killing of bacteria by tagging invaders through opsonisation, thereby promoting phagocytosis, and by direct lysis. Complement activity is routinely measured using functional assays that utilise erythrocytes as targets. The classical pathway haemolytic assay (CH50) with antibody sensitised sheep erythrocytes as target is used worldwide in clinical and research laboratories to measure complement activity in human and rodent sera. While there are no particular limitations in the human assay, measuring complement in mouse serum is more difficult and usually requires large amounts of serum, which is challenging to collect in experiments. In particular, it is challenging to measure the activities of individual mouse complement proteins. To overcome this hurdle, we have developed protocols that employ human sera depleted of single complement proteins as the source of the other complement proteins and test mouse serum to restore the relevant component. This simple haemolytic assay is a useful tool for confirming natural or engineered complement deficiencies and complement dysregulation in mouse models.
Collapse
Affiliation(s)
- Wioleta M Zelek
- Division of Infection and Immunity and Dementia Research Institute, School of Medicine, Cardiff University, Wales, United Kingdom
| |
Collapse
|
34
|
Niksirat H, Siino V, Steinbach C, Levander F. High-Resolution Proteomic Profiling Shows Sexual Dimorphism in Zebrafish Heart-Associated Proteins. J Proteome Res 2021; 20:4075-4088. [PMID: 34185526 DOI: 10.1021/acs.jproteome.1c00387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Understanding the molecular basis of sexual dimorphism in the cardiovascular system may contribute to the improvement of the outcome in biological, pharmacological, and toxicological studies as well as on the development of sex-based drugs and therapeutic approaches. Label-free protein quantification using high-resolution mass spectrometry was applied to detect sex-based proteome differences in the heart of zebrafish Danio rerio. Out of almost 3000 unique identified proteins in the heart, 79 showed significant abundance differences between male and female fish. The functional differences were mapped using enrichment analyses. Our results suggest that a large amount of materials needed for reproduction (e.g., sugars, lipids, proteins, etc.) may impose extra pressure on blood, vessels, and heart on their way toward the ovaries. In the present study, the female's heart shows a clear sexual dimorphism by changing abundance levels of numerous proteins, which could be a way to safely overcome material-induced elevated pressures. These proteins belong to the immune system, oxidative stress response, drug metabolization, detoxification, energy, metabolism, and so on. In conclusion, we showed that sex can induce dimorphism at the molecular level in nonsexual organs such as heart and must be considered as an important factor in cardiovascular research. Data are available via ProteomeXchange with identifier PXD023506.
Collapse
Affiliation(s)
- Hamid Niksirat
- Faculty of Fisheries and Protection of Waters, CENAKVA, University of South Bohemia in České Budějovice, Vodňany, 370 05 České Budějovice, Czech Republic
| | - Valentina Siino
- Department of Immunotechnology, Lund University, Lund 223 87, Sweden
| | - Christoph Steinbach
- Faculty of Fisheries and Protection of Waters, CENAKVA, University of South Bohemia in České Budějovice, Vodňany, 370 05 České Budějovice, Czech Republic
| | - Fredrik Levander
- Department of Immunotechnology, Lund University, Lund 223 87, Sweden.,National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Lund University, Lund 223 87, Sweden
| |
Collapse
|
35
|
Faranda AP, Shihan MH, Wang Y, Duncan MK. The effect of sex on the mouse lens transcriptome. Exp Eye Res 2021; 209:108676. [PMID: 34146586 DOI: 10.1016/j.exer.2021.108676] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 02/08/2023]
Abstract
The transcriptome of mammalian tissues differs between males and females, and these differences can change across the lifespan, likely regulating known sexual dimorphisms in disease prevalence and severity. Cataract, the most prevalent disease of the ocular lens, occurs at similar rates in young individuals, but its incidence is elevated in older women compared to men of the same age. However, the influence of sex on the lens transcriptome was unknown. RNAseq based transcriptomic profiling of young adult C57BL/6J mouse lens epithelial and fiber cells revealed that few genes are differentially expressed between the sexes. In contrast, lens cells from aged (24 month old) male and female C57BL/6J mice differentially expressed many genes, including several whose expression is lens preferred. Like cataracts, posterior capsular opacification (PCO), a major sequela of cataract surgery, may also be more prevalent in women. Lens epithelial cells isolated from mouse eyes 24 h after lens fiber cell removal exhibited numerous transcriptomic differences between the sexes, including genes implicated in complement cascades and extracellular matrix regulation, and these differences are much more pronounced in aged mice than in young mice. These results provide an unbiased basis for future studies on how sex affects the lens response to aging, cataract development, and cataract surgery.
Collapse
Affiliation(s)
- Adam P Faranda
- Department of Biological Sciences University of Delaware, Newark, DE, 19716, USA
| | - Mahbubul H Shihan
- Department of Biological Sciences University of Delaware, Newark, DE, 19716, USA
| | - Yan Wang
- Department of Biological Sciences University of Delaware, Newark, DE, 19716, USA
| | - Melinda K Duncan
- Department of Biological Sciences University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
36
|
Zhang H, Tang Y, Tao J. Sex-Related Overactivation of NLRP3 Inflammasome Increases Lethality of the Male COVID-19 Patients. Front Mol Biosci 2021; 8:671363. [PMID: 34150848 PMCID: PMC8212049 DOI: 10.3389/fmolb.2021.671363] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/20/2021] [Indexed: 12/30/2022] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2 infection, remains a dramatic threat to human life and economic well-being worldwide. Significant heterogeneity in the severity of disease was observed for patients infected with SARS-CoV-2 ranging from asymptomatic to severe cases. Moreover, male patients had a higher probability of suffering from high mortality and severe symptoms linked to cytokine storm and excessive inflammation. The NLRP3 inflammasome is presumably critical to this process. Sex differences may directly affect the activation of NLRP3 inflammasome, impacting the severity of observed COVID-19 symptoms. To elucidate the potential mechanisms underlying sex based differences in NLRP3 activation during SARS-CoV-2 infection, this review summarizes the reported mechanisms and identifies potential therapeutic targets.
Collapse
Affiliation(s)
| | | | - Jinhui Tao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
37
|
Mohammed Y, Michaud SA, Pětrošová H, Yang J, Ganguly M, Schibli D, Flenniken AM, Nutter LMJ, Adissu HA, Lloyd KCK, McKerlie C, Borchers CH. Proteotyping of knockout mouse strains reveals sex- and strain-specific signatures in blood plasma. NPJ Syst Biol Appl 2021; 7:25. [PMID: 34050187 PMCID: PMC8163790 DOI: 10.1038/s41540-021-00184-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 04/25/2021] [Indexed: 11/24/2022] Open
Abstract
We proteotyped blood plasma from 30 mouse knockout strains and corresponding wild-type mice from the International Mouse Phenotyping Consortium. We used targeted proteomics with internal standards to quantify 375 proteins in 218 samples. Our results provide insights into the manifested effects of each gene knockout at the plasma proteome level. We first investigated possible contamination by erythrocytes during sample preparation and labeled, in one case, up to 11 differential proteins as erythrocyte originated. Second, we showed that differences in baseline protein abundance between female and male mice were evident in all mice, emphasizing the necessity to include both sexes in basic research, target discovery, and preclinical effect and safety studies. Next, we identified the protein signature of each gene knockout and performed functional analyses for all knockout strains. Further, to demonstrate how proteome analysis identifies the effect of gene deficiency beyond traditional phenotyping tests, we provide in-depth analysis of two strains, C8a-/- and Npc2+/-. The proteins encoded by these genes are well-characterized providing good validation of our method in homozygous and heterozygous knockout mice. Ig alpha chain C region, a poorly characterized protein, was among the differentiating proteins in C8a-/-. In Npc2+/- mice, where histopathology and traditional tests failed to differentiate heterozygous from wild-type mice, our data showed significant difference in various lysosomal storage disease-related proteins. Our results demonstrate how to combine absolute quantitative proteomics with mouse gene knockout strategies to systematically study the effect of protein absence. The approach used here for blood plasma is applicable to all tissue protein extracts.
Collapse
Affiliation(s)
- Yassene Mohammed
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada.
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands.
| | - Sarah A Michaud
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada.
| | - Helena Pětrošová
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada
| | - Juncong Yang
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada
| | - Milan Ganguly
- The Center for Phenogenomics, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - David Schibli
- University of Victoria-Genome BC Proteomics Centre, Victoria, BC, Canada
| | - Ann M Flenniken
- The Center for Phenogenomics, Toronto, ON, Canada
- Sinai Health Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - Lauryl M J Nutter
- The Center for Phenogenomics, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | | | - K C Kent Lloyd
- Department of Surgery, School of Medicine, and Mouse Biology Program, University of California, Davis, CA, USA
| | | | - Christoph H Borchers
- Proteomics Centre, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada.
- Gerald Bronfman Department of Oncology, Jewish General Hospital, Montreal, QC, Canada.
- Department of Data Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Skolkovo Innovation Center, Moscow, Russia.
| |
Collapse
|
38
|
Wu M, Rowe JM, Fleming SD. Complement Initiation Varies by Sex in Intestinal Ischemia Reperfusion Injury. Front Immunol 2021; 12:649882. [PMID: 33868287 PMCID: PMC8047102 DOI: 10.3389/fimmu.2021.649882] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/08/2021] [Indexed: 01/03/2023] Open
Abstract
Intestinal ischemia reperfusion (IR)-induced tissue injury represents an acute inflammatory response with significant morbidity and mortality. The mechanism of IR-induced injury is not fully elucidated, but recent studies suggest a critical role for complement activation and for differences between sexes. To test the hypothesis that complement initiation differs by sex in intestinal IR, we performed intestinal IR on male and female WT C57B6L/, C1q-/-, MBL-/-, or properdin (P)-/- mice. Intestinal injury, C3b and C5a production and ex vivo secretions were analyzed. Initial studies demonstrated a difference in complement mRNA and protein in male and female WT mice. In response to IR, male C1q-, MBL- and P-deficient mice sustained less injury than male WT mice. In contrast, only female MBL-/- mice sustained significantly less injury than female wildtype mice. Importantly, wildtype, C1q-/- and P-/- female mice sustained significant less injury than the corresponding male mice. In addition, both C1q and MBL expression and deposition increased in WT male mice, while only elevated MBL expression and deposition occurred in WT female mice. These data suggested that males use both C1q and MBL pathways, while females tend to depend on lectin pathway during intestinal IR. Females produced significantly less serum C5a in MBL-/- and P-/- mice. Our findings suggested that complement activation plays a critical role in intestinal IR in a sex-dependent manner.
Collapse
Affiliation(s)
- Miaomiao Wu
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Jennifer M. Rowe
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Sherry D. Fleming
- Division of Biology, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
39
|
Ricardo-da-Silva FY, Armstrong-Jr R, Vidal-dos-Santos M, Correia CDJ, Coutinho e Silva RDS, da Anunciação LF, Moreira LFP, Leuvenink HGD, Breithaupt-Faloppa AC. Long-term lung inflammation is reduced by estradiol treatment in brain dead female rats. Clinics (Sao Paulo) 2021; 76:e3042. [PMID: 34406272 PMCID: PMC8341046 DOI: 10.6061/clinics/2021/e3042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 06/28/2021] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVES Lung transplantation is limited by the systemic repercussions of brain death (BD). Studies have shown the potential protective role of 17β-estradiol on the lungs. Here, we aimed to investigate the effect of estradiol on the long-lasting lung inflammatory state to understand a possible therapeutic application in lung donors with BD. METHODS Female Wistar rats were separated into 3 groups: BD, subjected to brain death (6h); E2-T0, treated with 17β-estradiol (50 μg/mL, 2 mL/h) immediately after brain death; and E2-T3, treated with 17β-estradiol (50 μg/ml, 2 ml/h) after 3h of BD. Complement system activity and macrophage presence were analyzed. TNF-α, IL-1β, IL-10, and IL-6 gene expression (RT-PCR) and levels in 24h lung culture medium were quantified. Finally, analysis of caspase-3 gene and protein expression in the lung was performed. RESULTS Estradiol reduced complement C3 protein and gene expression. The presence of lung macrophages was not modified by estradiol, but the release of inflammatory mediators was reduced and TNF-α and IL-1β gene expression were reduced in the E2-T3 group. In addition, caspase-3 protein expression was reduced by estradiol in the same group. CONCLUSIONS Brain death-induced lung inflammation in females is modulated by estradiol treatment. Study data suggest that estradiol can control the inflammatory response by modulating the release of mediators after brain death in the long term. These results strengthen the idea of estradiol as a therapy for donor lungs and improving transplant outcomes.
Collapse
Affiliation(s)
- Fernanda Yamamoto Ricardo-da-Silva
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Roberto Armstrong-Jr
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Marina Vidal-dos-Santos
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Cristiano de Jesus Correia
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Raphael dos Santos Coutinho e Silva
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Lucas Ferreira da Anunciação
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Luiz Felipe Pinho Moreira
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | | | - Ana Cristina Breithaupt-Faloppa
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Corresponding author. E-mail:
| |
Collapse
|
40
|
Baiardo Redaelli M, Landoni G, Di Napoli D, Morselli F, Sartorelli M, Sartini C, Ruggeri A, Salonia A, Dagna L, Zangrillo A. Novel Coronavirus Disease (COVID-19) in Italian Patients: Gender Differences in Presentation and Severity. SAUDI JOURNAL OF MEDICINE & MEDICAL SCIENCES 2021; 9:59-62. [PMID: 33519345 PMCID: PMC7839571 DOI: 10.4103/sjmms.sjmms_542_20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/06/2020] [Accepted: 11/19/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND In the first wave of the novel coronavirus (severe acute respiratory syndrome coronavirus 2) infections, Italy experienced a heavy burden of hospital admissions for acute respiratory distress syndromes associated with the novel coronavirus disease (COVID-19). Early evidence suggested that females are less affected than males. OBJECTIVE This study aimed to assess the gender-related differences in presentation and severity among COVID-19 patients admitted to IRCCS San Raffaele Hospital, Milan, Italy. MATERIALS AND METHODS This prospective observational study included all patients admitted to the hospital between February 25 and April 19, 2020, with a positive real-time reverse-transcriptase polymerase chain reaction for COVID-19. The following data were collected: date of admission, gender, age and details of intensive care unit admission and outcomes. RESULTS A total of 901 patients with COVID-19 were admitted to the hospital and provided consent for the study. Of these, 284 were female (31.5%). The percentage of admitted female patients significantly increased over time (25.9% of all admissions in the first half of the study period vs. 37.1% in the second half; P < 0.001). Females accounted for 14.4% of all COVID-19 intensive care unit admissions. There was no gender-based difference in the overall hospital mortality: 20.1% for females and 19.2% for males (P = 0.8). CONCLUSIONS In our hospital, which was in the epicenter of the first wave of COVID-19 pandemic in Italy, female patients were few, presented late and were less critical than male patients.
Collapse
Affiliation(s)
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Hospital, Milan, Italy
- Department of Faculty of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Davide Di Napoli
- Department of Health Directorate, IRCCS San Raffaele Hospital, Milan, Italy
| | - Federica Morselli
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Hospital, Milan, Italy
| | - Marianna Sartorelli
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Hospital, Milan, Italy
| | - Chiara Sartini
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Hospital, Milan, Italy
| | - Annalisa Ruggeri
- Department of Hematology and Stem Cell Transplantation, IRCCS Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Salonia
- Department of Faculty of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Lorenzo Dagna
- Department of Faculty of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Milan, Italy
| | - Alberto Zangrillo
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Hospital, Milan, Italy
- Department of Faculty of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
41
|
Sachs S, Niu L, Geyer P, Jall S, Kleinert M, Feuchtinger A, Stemmer K, Brielmeier M, Finan B, DiMarchi RD, Tschöp MH, Wewer Albrechtsen N, Mann M, Müller TD, Hofmann SM. Plasma proteome profiles treatment efficacy of incretin dual agonism in diet-induced obese female and male mice. Diabetes Obes Metab 2021; 23:195-207. [PMID: 33001570 DOI: 10.1111/dom.14215] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/21/2020] [Accepted: 09/26/2020] [Indexed: 02/06/2023]
Abstract
AIMS Unimolecular peptides targeting the receptors for glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) (GLP-1/GIP co-agonist) have been shown to outperform each single peptide in the treatment of obesity and cardiometabolic disease in preclinical and clinical trials. By combining physiological treatment endpoints with plasma proteomic profiling (PPP), we aimed to identify biomarkers to advance non-invasive metabolic monitoring of compound treatment success and exploration of ulterior treatment effects on an individual basis. MATERIALS AND METHODS We performed metabolic phenotyping along with PPP in body weight-matched male and female diet-induced obese (DIO) mice treated for 21 days with phosphate-buffered saline, single GIP and GLP-1 mono-agonists, or a GLP-1/GIP co-agonist. RESULTS GLP-1R/GIPR co-agonism improved obesity, glucose intolerance, non-alcoholic fatty liver disease (NAFLD) and dyslipidaemia with superior efficacy in both male and female mice compared with mono-agonist treatments. PPP revealed broader changes of plasma proteins after GLP-1/GIP co-agonist compared with mono-agonist treatments in both sexes, including established and potential novel biomarkers for systemic inflammation, NAFLD and atherosclerosis. Subtle sex-specific differences have been observed in metabolic phenotyping and PPP. CONCLUSIONS We herein show that a recently developed unimolecular GLP-1/GIP co-agonist is more efficient in improving metabolic disease than either mono-agonist in both sexes. PPP led to the identification of a sex-independent protein panel with the potential to monitor non-invasively the treatment efficacies on metabolic function of this clinically advancing GLP-1/GIP co-agonist.
Collapse
Affiliation(s)
- Stephan Sachs
- Institute for Diabetes and Regeneration, Helmholtz Diabetes Center at Helmholtz Centre Munich, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - Lili Niu
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Philipp Geyer
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Sigrid Jall
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - Maximilian Kleinert
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Neuherberg, Germany
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Center Munich, Neuherberg, Germany
| | - Kerstin Stemmer
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Markus Brielmeier
- Helmholtz Zentrum München-German Research Center for Environmental Health, Research Unit Comparative Medicine, Neuherberg, Germany
| | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, Indiana
| | - Richard D DiMarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, Indiana
- Department of Chemistry, Indiana University, Bloomington, Indiana
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Neuherberg, Germany
- Division of Metabolic Diseases, Technische Universität München, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Nicolai Wewer Albrechtsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Matthias Mann
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany
| | - Susanna M Hofmann
- Institute for Diabetes and Regeneration, Helmholtz Diabetes Center at Helmholtz Centre Munich, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV, Klinikum der LMU, Munich, Germany
| |
Collapse
|
42
|
Malsy J, Alvarado AC, Lamontagne JO, Strittmatter K, Marneros AG. Distinct effects of complement and of NLRP3- and non-NLRP3 inflammasomes for choroidal neovascularization. eLife 2020; 9:60194. [PMID: 33305736 PMCID: PMC7732340 DOI: 10.7554/elife.60194] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 11/05/2020] [Indexed: 12/16/2022] Open
Abstract
NLRP3 inflammasome activation and complement-mediated inflammation have been implicated in promoting choroidal neovascularization (CNV) in age-related macular degeneration (AMD), but central questions regarding their contributions to AMD pathogenesis remain unanswered. Key open questions are (1) whether NLRP3 inflammasome activation mainly in retinal pigment epithelium (RPE) or rather in non-RPE cells promotes CNV, (2) whether inflammasome activation in CNV occurs via NLRP3 or also through NLRP3-independent mechanisms, and (3) whether complement activation induces inflammasome activation in CNV. Here we show in a neovascular AMD mouse model that NLRP3 inflammasome activation in non-RPE cells but not in RPE cells promotes CNV. We demonstrate that both NLRP3-dependent and NLRP3-independent inflammasome activation mechanisms induce CNV. Finally, we find that complement and inflammasomes promote CNV through independent mechanisms. Our findings uncover an unexpected role of non-NLRP3 inflammasomes for CNV and suggest that combination therapies targeting inflammasomes and complement may offer synergistic benefits to inhibit CNV.
Collapse
Affiliation(s)
- Jakob Malsy
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, United States.,Department of Ophthalmology, University of Halle, Halle, Germany
| | - Andrea C Alvarado
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, United States
| | - Joseph O Lamontagne
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, United States
| | - Karin Strittmatter
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, United States
| | - Alexander G Marneros
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, United States
| |
Collapse
|
43
|
Bevan RJ, Hughes TR, Williams PA, Good MA, Morgan BP, Morgan JE. Retinal ganglion cell degeneration correlates with hippocampal spine loss in experimental Alzheimer's disease. Acta Neuropathol Commun 2020; 8:216. [PMID: 33287900 PMCID: PMC7720390 DOI: 10.1186/s40478-020-01094-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023] Open
Abstract
Neuronal dendritic and synaptic pruning are early features of neurodegenerative diseases, including Alzheimer's disease. In addition to brain pathology, amyloid plaque deposition, microglial activation, and cell loss occur in the retinas of human patients and animal models of Alzheimer's disease. Retinal ganglion cells, the output neurons of the retina, are vulnerable to damage in neurodegenerative diseases and are a potential opportunity for non-invasive clinical diagnosis and monitoring of Alzheimer's progression. However, the extent of retinal involvement in Alzheimer's models and how well this reflects brain pathology is unclear. Here we have quantified changes in retinal ganglion cells dendritic structure and hippocampal dendritic spines in three well-studied Alzheimer's mouse models, Tg2576, 3xTg-AD and APPNL-G-F. Dendritic complexity of DiOlistically labelled retinal ganglion cells from retinal explants was reduced in all three models in an age-, gender-, and receptive field-dependent manner. DiOlistically labelled hippocampal slices showed spine loss in CA1 apical dendrites in all three Alzheimer's models, mirroring the early stages of neurodegeneration as seen in the retina. Morphological classification showed that loss of thin spines predominated in all. The demonstration that retinal ganglion cells dendritic field reduction occurs in parallel with hippocampal dendritic spine loss in all three Alzheimer's models provide compelling support for the use of retinal neurodegeneration. As retinal dendritic changes are within the optical range of current clinical imaging systems (for example optical coherence tomography), our study makes a case for imaging the retina as a non-invasive way to diagnose disease and monitor progression in Alzheimer's disease.
Collapse
Affiliation(s)
- Ryan J Bevan
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, CF24 4HQ, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Tim R Hughes
- UK Dementia Research Institute, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Mark A Good
- School of Psychology, Cardiff University, Cardiff, UK
| | - B Paul Morgan
- UK Dementia Research Institute, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - James E Morgan
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
44
|
Pedersen H, Jensen RK, Jensen JMB, Fox R, Pedersen DV, Olesen HG, Hansen AG, Christiansen D, Mazarakis SMM, Lojek N, Hansen P, Gadeberg TAF, Zarantonello A, Laursen NS, Mollnes TE, Johnson MB, Stevens B, Thiel S, Andersen GR. A Complement C3-Specific Nanobody for Modulation of the Alternative Cascade Identifies the C-Terminal Domain of C3b as Functional in C5 Convertase Activity. THE JOURNAL OF IMMUNOLOGY 2020; 205:2287-2300. [PMID: 32938727 DOI: 10.4049/jimmunol.2000752] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/13/2020] [Indexed: 12/17/2022]
Abstract
The complement system is an intricate cascade of the innate immune system and plays a key role in microbial defense, inflammation, organ development, and tissue regeneration. There is increasing interest in developing complement regulatory and inhibitory agents to treat complement dysfunction. In this study, we describe the nanobody hC3Nb3, which is specific for the C-terminal C345c domain of human and mouse complement component C3/C3b/C3c and potently inhibits C3 cleavage by the alternative pathway. A high-resolution structure of the hC3Nb3-C345c complex explains how the nanobody blocks proconvertase assembly. Surprisingly, although the nanobody does not affect classical pathway-mediated C3 cleavage, hC3Nb3 inhibits classical pathway-driven hemolysis, suggesting that the C-terminal domain of C3b has an important function in classical pathway C5 convertase activity. The hC3Nb3 nanobody binds C3 with low nanomolar affinity in an SDS-resistant complex, and the nanobody is demonstrated to be a powerful reagent for C3 detection in immunohistochemistry and flow cytometry. Overall, the hC3Nb3 nanobody represents a potent inhibitor of both the alternative pathway and the terminal pathway, with possible applications in complement research, diagnostics, and therapeutics.
Collapse
Affiliation(s)
- Henrik Pedersen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus, Denmark
| | - Rasmus K Jensen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus, Denmark
| | | | - Rachel Fox
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Dennis V Pedersen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus, Denmark
| | - Heidi G Olesen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus, Denmark
| | - Annette G Hansen
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
| | | | - Sofia M M Mazarakis
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus, Denmark
| | - Neal Lojek
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Pernille Hansen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus, Denmark
| | - Trine A F Gadeberg
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus, Denmark
| | | | - Nick S Laursen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus, Denmark
| | - Tom Eirik Mollnes
- Research Laboratory, Nordland Hospital, 8092 Bodø, Norway.,K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, 9037 Tromsø, Norway.,Department of Immunology, Oslo University Hospital, University of Oslo, 0318 Oslo, Norway.,Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7491 Trondheim, Norway; and
| | - Matthew B Johnson
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142.,Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115
| | - Beth Stevens
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142.,Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115
| | - Steffen Thiel
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
| | - Gregers R Andersen
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus, Denmark;
| |
Collapse
|
45
|
Hammond JW, Bellizzi MJ, Ware C, Qiu WQ, Saminathan P, Li H, Luo S, Ma SA, Li Y, Gelbard HA. Complement-dependent synapse loss and microgliosis in a mouse model of multiple sclerosis. Brain Behav Immun 2020; 87:739-750. [PMID: 32151684 PMCID: PMC8698220 DOI: 10.1016/j.bbi.2020.03.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory, neurodegenerative disease of the CNS characterized by both grey and white matter injury. Microglial activation and a reduction in synaptic density are key features of grey matter pathology that can be modeled with MOG35-55 experimental autoimmune encephalomyelitis (EAE). Complement deposition combined with microglial engulfment has been shown during normal development and in disease as a mechanism for pruning synapses. We tested whether there is excess complement production in the EAE hippocampus and whether complement-dependent synapse loss is a source of degeneration in EAE using C1qa and C3 knockout mice. We found that C1q and C3 protein and mRNA levels were elevated in EAE mice. Genetic loss of C3 protected mice from EAE-induced synapse loss, reduced microglial activation, decreased the severity of the EAE clinical score, and protected memory/freezing behavior after contextual fear conditioning. C1qa KO mice with EAE showed little to no change on these measurements compared to WT EAE mice. Thus, pathologic expression and activation of the early complement pathway, specifically at the level of C3, contributes to hippocampal grey matter pathology in the EAE.
Collapse
Affiliation(s)
- Jennetta W. Hammond
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642.,Correspondence: Jennetta W. Hammond,
University of Rochester, Center for Neurotherapeutics Discovery, 601 Elmwood
Avenue, Box 645, Rochester, NY 14642, USA,
, Phone:
1-585-273-2872
| | - Matthew J. Bellizzi
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neuroscience, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Caroline Ware
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Wen Q. Qiu
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Priyanka Saminathan
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Microbiology and Immunology, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Herman Li
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Shaopeiwen Luo
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Stefanie A. Ma
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Yuanhao Li
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642
| | - Harris A. Gelbard
- Center for Neurotherapeutics Discovery, University of
Rochester Medical Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neurology, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642.,Department of Neuroscience, University of Rochester Medical
Center, 601 Elmwood Avenue, Rochester NY 14642
| |
Collapse
|
46
|
Pedersen H, Jensen RK, Hansen AG, Gadeberg TAF, Thiel S, Laursen NS, Andersen GR. A C3-specific nanobody that blocks all three activation pathways in the human and murine complement system. J Biol Chem 2020; 295:8746-8758. [PMID: 32376685 DOI: 10.1074/jbc.ra119.012339] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/28/2020] [Indexed: 12/18/2022] Open
Abstract
The complement system is a tightly controlled proteolytic cascade in the innate immune system, which tags intruding pathogens and dying host cells for clearance. An essential protein in this process is complement component C3. Uncontrolled complement activation has been implicated in several human diseases and disorders and has spurred the development of therapeutic approaches that modulate the complement system. Here, using purified proteins and several biochemical assays and surface plasmon resonance, we report that our nanobody, hC3Nb2, inhibits C3 deposition by all complement pathways. We observe that the hC3Nb2 nanobody binds human native C3 and its degradation products with low nanomolar affinity and does not interfere with the endogenous regulation of C3b deposition mediated by Factors H and I. Using negative stain EM analysis and functional assays, we demonstrate that hC3Nb2 inhibits the substrate-convertase interaction by binding to the MG3 and MG4 domains of C3 and C3b. Furthermore, we notice that hC3Nb2 is cross-reactive and inhibits the lectin and alternative pathway in murine serum. We conclude that hC3Nb2 is a potent, general, and versatile inhibitor of the human and murine complement cascades. Its cross-reactivity suggests that this nanobody may be valuable for analysis of complement activation within animal models of both acute and chronic diseases.
Collapse
Affiliation(s)
- Henrik Pedersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Rasmus K Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Trine A F Gadeberg
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Steffen Thiel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Nick S Laursen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Gregers R Andersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
47
|
Krukonis ES, Thomson JJ. Complement evasion mechanisms of the systemic pathogens Yersiniae and Salmonellae. FEBS Lett 2020; 594:2598-2620. [DOI: 10.1002/1873-3468.13771] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 01/12/2023]
Affiliation(s)
- Eric S. Krukonis
- Division of Integrated Biomedical Sciences University of Detroit Mercy School of Dentistry Detroit MI USA
| | - Joshua J. Thomson
- Division of Integrated Biomedical Sciences University of Detroit Mercy School of Dentistry Detroit MI USA
| |
Collapse
|
48
|
Abstract
The recognition of microbial or danger-associated molecular patterns by complement proteins initiates a cascade of events that culminates in the activation of surface complement receptors on immune cells. Such signalling pathways converge with those activated downstream of pattern recognition receptors to determine the type and magnitude of the immune response. Intensive investigation in the field has uncovered novel pathways that link complement-mediated signalling with homeostatic and pathological T cell responses. More recently, the observation that complement proteins also act in the intracellular space to shape T cell fates has added a new layer of complexity. Here, we consider fundamental mechanisms and novel concepts at the interface of complement biology and immunity and discuss how these affect the maintenance of homeostasis and the development of human pathology.
Collapse
|
49
|
Brosseron F, Kolbe C, Santarelli F, Carvalho S, Antonell A, Castro‐Gomez S, Tacik P, Namasivayam AA, Mangone G, Schneider R, Latz E, Wüllner U, Svenningsson P, Sánchez‐Valle R, Molinuevo JL, Corvol J, Heneka MT. Multicenter Alzheimer's and Parkinson's disease immune biomarker verification study. Alzheimers Dement 2020; 16:292-304. [DOI: 10.1016/j.jalz.2019.07.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Frederic Brosseron
- Department of Neurodegenerative Diseases & Geropsychiatry/Neurology University of Bonn Medical Center Bonn Germany
- German Center for Neurodegenerative Diseases (DZNE) Bonn Germany
| | | | - Francesco Santarelli
- Department of Neurodegenerative Diseases & Geropsychiatry/Neurology University of Bonn Medical Center Bonn Germany
- German Center for Neurodegenerative Diseases (DZNE) Bonn Germany
| | - Stephanie Carvalho
- Institut du Cerveau et de la Moelle épinière, Sorbonne Université, Assistance‐Publique Hôpitaux de Paris, INSERM, UMRS 1127, CNRS, UMR 7225, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié‐Salpêtrière, Department of Neurology, Centre d'Investigation Clinique Neurosciences Paris France
| | - Anna Antonell
- Alzheimer's disease and other cognitive disorders Unit, Hospital Clínic, Fundació Clínic per a la Recerca Biomèdica Institut d'Investigacions Biomè; diques August Pi i Sunyer (IDIBAPS) Barcelona Spain
| | - Sergio Castro‐Gomez
- Department of Neurodegenerative Diseases & Geropsychiatry/Neurology University of Bonn Medical Center Bonn Germany
| | - Pawel Tacik
- Department of Neurodegenerative Diseases & Geropsychiatry/Neurology University of Bonn Medical Center Bonn Germany
| | - Aishwarya Alex Namasivayam
- Luxembourg Centre for Systems Biomedicine (LCSB) University of Luxembourg Campus Belval Belvaux Luxembourg
| | - Graziella Mangone
- Institut du Cerveau et de la Moelle épinière, Sorbonne Université, Assistance‐Publique Hôpitaux de Paris, INSERM, UMRS 1127, CNRS, UMR 7225, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié‐Salpêtrière, Department of Neurology, Centre d'Investigation Clinique Neurosciences Paris France
| | - Reinhard Schneider
- Luxembourg Centre for Systems Biomedicine (LCSB) University of Luxembourg Campus Belval Belvaux Luxembourg
| | - Eicke Latz
- German Center for Neurodegenerative Diseases (DZNE) Bonn Germany
- University of Bonn Medical Center Institute of Innate Immune Bonn Germany
| | - Ullrich Wüllner
- German Center for Neurodegenerative Diseases (DZNE) Bonn Germany
- Department of Neurology University of Bonn Medical Center Bonn Germany
| | - Per Svenningsson
- Department of Clinical Neuroscience Karolinska Institutet Stockholm Sweden
| | - Raquel Sánchez‐Valle
- Alzheimer's disease and other cognitive disorders Unit, Hospital Clínic, Fundació Clínic per a la Recerca Biomèdica Institut d'Investigacions Biomè; diques August Pi i Sunyer (IDIBAPS) Barcelona Spain
| | - José Luis Molinuevo
- Barcelonabeta Brain Research Center, Pasqual Maragall Foundation Barcelona Spain
| | - Jean‐Christophe Corvol
- Institut du Cerveau et de la Moelle épinière, Sorbonne Université, Assistance‐Publique Hôpitaux de Paris, INSERM, UMRS 1127, CNRS, UMR 7225, Institut du Cerveau et de la Moelle épinière, ICM, Hôpital Pitié‐Salpêtrière, Department of Neurology, Centre d'Investigation Clinique Neurosciences Paris France
| | - Michael T. Heneka
- Department of Neurodegenerative Diseases & Geropsychiatry/Neurology University of Bonn Medical Center Bonn Germany
- German Center for Neurodegenerative Diseases (DZNE) Bonn Germany
| | | |
Collapse
|
50
|
Schein TN, Blackburn TE, Heath SL, Barnum SR. Plasma levels of soluble membrane attack complex are elevated despite viral suppression in HIV patients with poor immune reconstitution. Clin Exp Immunol 2019; 198:359-366. [PMID: 31461782 PMCID: PMC6857077 DOI: 10.1111/cei.13366] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2019] [Indexed: 12/22/2022] Open
Abstract
The complement system is now a therapeutic target for the management of serious and life-threatening conditions such as paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, glomerulonephritis and other diseases caused by complement deficiencies or genetic variants. As complement therapeutics expand into more clinical conditions, monitoring complement activation is increasingly important, as is the baseline levels of complement activation fragments in blood or other body fluid levels. Although baseline complement levels have been reported in the literature, the majority of these data were generated using non-standard assays and with variable sample handling, potentially skewing results. In this study, we examined the plasma and serum levels of the soluble membrane attack complex of complement (sMAC). sMAC is formed in the fluid phase when complement is activated through the terminal pathway. It binds the regulatory proteins vitronectin and/or clusterin and cannot insert into cell membranes, and can serve as a soluble diagnostic marker in infectious disease settings, as previously shown for intraventricular shunt infections. Here we show that in healthy adults, serum sMAC levels were significantly higher than those in plasma, that plasma sMAC levels were similar between in African Americans and Caucasians and that plasma sMAC levels increase with age. Plasma sMAC levels were significantly higher in virally suppressed people living with HIV (PLWH) compared to non-HIV infected healthy donors. More specifically, PLWH with CD4+ T cell counts below 200 had even greater sMAC levels, suggesting diagnostic value in monitoring sMAC levels in this group.
Collapse
Affiliation(s)
- T. N. Schein
- Department of MicrobiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - T. E. Blackburn
- Department of MicrobiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - S. L. Heath
- Department of Medicine, Division of Infectious DiseaseUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - S. R. Barnum
- Department of MicrobiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|