Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ganesh Kolumam is active.

Publication


Featured researches published by Ganesh Kolumam.


Nature | 2014

Interleukin-22 alleviates metabolic disorders and restores mucosal immunity in diabetes

Xiaoting Wang; Naruhisa Ota; Paolo Manzanillo; Lance Kates; Jose Zavala-Solorio; Céline Eidenschenk; Juan Zhang; Justin Lesch; Wyne P. Lee; Jed Ross; Lauri Diehl; Nicholas van Bruggen; Ganesh Kolumam; Wenjun Ouyang

The connection between an altered gut microbiota and metabolic disorders such as obesity, diabetes, and cardiovascular disease is well established. Defects in preserving the integrity of the mucosal barriers can result in systemic endotoxaemia that contributes to chronic low-grade inflammation, which further promotes the development of metabolic syndrome. Interleukin (IL)-22 exerts essential roles in eliciting antimicrobial immunity and maintaining mucosal barrier integrity within the intestine. Here we investigate the connection between IL-22 and metabolic disorders. We find that the induction of IL-22 from innate lymphoid cells and CD4+ T cells is impaired in obese mice under various immune challenges, especially in the colon during infection with Citrobacter rodentium. While innate lymphoid cell populations are largely intact in obese mice, the upregulation of IL-23, a cytokine upstream of IL-22, is compromised during the infection. Consequently, these mice are susceptible to C. rodentium infection, and both exogenous IL-22 and IL-23 are able to restore the mucosal host defence. Importantly, we further unveil unexpected functions of IL-22 in regulating metabolism. Mice deficient in IL-22 receptor and fed with high-fat diet are prone to developing metabolic disorders. Strikingly, administration of exogenous IL-22 in genetically obese leptin-receptor-deficient (db/db) mice and mice fed with high-fat diet reverses many of the metabolic symptoms, including hyperglycaemia and insulin resistance. IL-22 shows diverse metabolic benefits, as it improves insulin sensitivity, preserves gut mucosal barrier and endocrine functions, decreases endotoxaemia and chronic inflammation, and regulates lipid metabolism in liver and adipose tissues. In summary, we identify the IL-22 pathway as a novel target for therapeutic intervention in metabolic diseases.


Nature Medicine | 2009

Targeted depletion of lymphotoxin-alpha-expressing TH1 and TH17 cells inhibits autoimmune disease.

Eugene Y. Chiang; Ganesh Kolumam; Xin Yu; Michelle Francesco; Sinisa Ivelja; Ivan Peng; Peter Gribling; Jean Shu; Wyne P. Lee; Canio J. Refino; Mercedesz Balazs; Andres Paler-Martinez; Allen Nguyen; Judy Young; Kai H. Barck; Richard A. D. Carano; Ron Ferrando; Lauri Diehl; Devavani Chatterjea; Jane L. Grogan

Uncontrolled T helper type 1 (TH1) and TH17 cells are associated with autoimmune responses. We identify surface lymphotoxin-α (LT-α) as common to TH0, TH1 and TH17 cells and employ a unique strategy to target these subsets using a depleting monoclonal antibody (mAb) directed to surface LT-α. Depleting LT-α–specific mAb inhibited T cell–mediated models of delayed-type hypersensitivity and experimental autoimmune encephalomyelitis. In collagen-induced arthritis (CIA), preventive and therapeutic administration of LT-α–specific mAb inhibited disease, and immunoablated T cells expressing interleukin-17 (IL-17), interferon-γ and tumor necrosis factor-α (TNF-α), whereas decoy lymphotoxin-β receptor (LT-βR) fusion protein had no effect. A mutation in the Fc tail, rendering the antibody incapable of Fcγ receptor binding and antibody-dependent cellular cytotoxicity activity, abolished all in vivo effects. Efficacy in CIA was preceded by a loss of rheumatoid-associated cytokines IL-6, IL-1β and TNF-α within joints. These data indicate that depleting LT-α–expressing lymphocytes with LT-α–specific mAb may be beneficial in the treatment of autoimmune disease.


Cell | 2010

PlGF Blockade Does Not Inhibit Angiogenesis during Primary Tumor Growth

Carlos Bais; Xiumin Wu; Jenny Yao; Suya Yang; Yongping Crawford; Krista McCutcheon; Christine Tan; Ganesh Kolumam; Jean-Michel Vernes; Jeffrey Eastham-Anderson; Peter Haughney; Marcin Kowanetz; Thijs J. Hagenbeek; Ian Kasman; Hani Bou Reslan; Jed Ross; Nick van Bruggen; Richard A. D. Carano; Yu-Ju Gloria Meng; Jo-Anne Hongo; Jean Philippe Stephan; Masabumi Shibuya; Napoleone Ferrara

It has been recently reported that treatment with an anti-placenta growth factor (PlGF) antibody inhibits metastasis and primary tumor growth. Here we show that, although anti-PlGF treatment inhibited wound healing, extravasation of B16F10 cells, and growth of a tumor engineered to overexpress the PlGF receptor (VEGFR-1), neutralization of PlGF using four novel blocking antibodies had no significant effect on tumor angiogenesis in 15 models. Also, genetic ablation of the tyrosine kinase domain of VEGFR-1 in the host did not result in growth inhibition of the anti-VEGF-A sensitive or resistant tumors tested. Furthermore, combination of anti-PlGF with anti-VEGF-A antibodies did not result in greater antitumor efficacy than anti-VEGF-A monotherapy. In conclusion, our data argue against an important role of PlGF during primary tumor growth in most models and suggest that clinical evaluation of anti-PlGF antibodies may be challenging.


Science Translational Medicine | 2011

Amelioration of Type 2 Diabetes by Antibody-Mediated Activation of Fibroblast Growth Factor Receptor 1

Ai-Luen Wu; Ganesh Kolumam; Scott Stawicki; Yongmei Chen; Jun Li; Jose Zavala-Solorio; Khanhky Phamluong; Bo Feng; Li Li; Scot A. Marsters; Lance Kates; Nicholas van Bruggen; Maya Leabman; Anne Wong; David West; Howard M. Stern; Elizabeth Luis; Hok Seon Kim; Daniel G. Yansura; Andrew S. Peterson; Ellen Filvaroff; Yan Wu; Junichiro Sonoda

Antibody-mediated activation of fibroblast growth factor receptor 1 reverses the diabetic phenotype in mice, likely by affecting brown adipose tissues. Getting at Brown Fat It’s fun to indulge in holiday cheer, if only a holiday miracle allowed one to avoid the often-linked weight gain. At the molecular level, obesity and type 2 diabetes can be linked by the fibroblast growth factor (FGF) family of proteins and their receptors (FGFRs), with some factors showing disease-reversing capabilities. For instance, overweight, diabetic mice treated with FGF21 regain normal metabolism and lose weight, even without spending hours on a treadmill. However, attempts to use this fat-burning factor in humans have not been successful, owing to poor pharmacokinetics as well as concerns over negative effects of modified FGF21 proteins. In this issue, Wu and colleagues describe an antibody-based FGF21 mimic that circumvents these limitations to overcome metabolic disease in mice. The authors reasoned that robust drugs that closely mimic FGF21 function would similarly exert antidiabetic effects. Using phage display technology, Wu et al. identified monoclonal antibodies (R1MAbs) that were specifically targeted tissues that play key roles in diabetes and obesity, including adipose (fat) tissue. In contrast to FGF21, which binds several forms of the FGFR throughout the body, the phage-derived R1MAbs bound only to FGFR1—a receptor present in the pancreas and in brown and white adipose tissues. Diabetic mice with high blood sugar (hyperglycemia) were injected once with either R1MAbs or a control antibody. Within 1 week, blood glucose concentrations in the R1MAb-treated mice were normalized and remained at lower levels compared to placebo-treated mice for more than 1 month without reaching dangerously low blood glucose concentrations (hypoglycemia). The R1MAbs also helped the diabetic mice to lose weight, indicating that this antibody agonist of FGFR1 is a dual-action drug for both diabetes and obesity. Wu et al. also shed light on the mechanism of action of their R1MAbs, showing that they work via FGFR homodimerization in brown adipose tissue. With improved pharmacokinetics over FGF21, in addition to a specific receptor-targeting mechanism, these R1MAbs could enter human clinical trials for diabetes and other obesity-related diseases in the near future. Unfortunately, a miracle drug won’t be available in time for the holidays, so perhaps, this year, opt for the sugar-free egg nog. Clinical use of recombinant fibroblast growth factor 21 (FGF21) for the treatment of type 2 diabetes and other disorders linked to obesity has been proposed; however, its clinical development has been challenging owing to its poor pharmacokinetics. Here, we describe an alternative antidiabetic strategy using agonistic anti-FGFR1 (FGF receptor 1) antibodies (R1MAbs) that mimic the metabolic effects of FGF21. A single injection of R1MAb into obese diabetic mice induced acute and sustained amelioration of hyperglycemia, along with marked improvement in hyperinsulinemia, hyperlipidemia, and hepatosteatosis. R1MAb activated the mitogen-activated protein kinase pathway in adipose tissues, but not in liver, and neither FGF21 nor R1MAb improved glucose clearance in lipoatrophic mice, which suggests that adipose tissues played a central role in the observed metabolic effects. In brown adipose tissues, both FGF21 and R1MAb induced phosphorylation of CREB (cyclic adenosine 5′-monophosphate response element–binding protein), and mRNA expression of PGC-1α (peroxisome proliferator–activated receptor-γ coactivator 1α) and the downstream genes associated with oxidative metabolism. Collectively, we propose FGFR1 in adipose tissues as a major functional receptor for FGF21, as an upstream regulator of PGC-1α, and as a compelling target for antibody-based therapy for type 2 diabetes and other obesity-associated disorders.


Nature | 2016

Phosphorylation and linear ubiquitin direct A20 inhibition of inflammation

Ingrid E. Wertz; Kim Newton; Dhaya Seshasayee; Saritha Kusam; Cynthia Lam; Juan Zhang; Nataliya Popovych; Elizabeth Helgason; Allyn J. Schoeffler; Surinder Jeet; Nandhini Ramamoorthi; Lorna Kategaya; Robert J. Newman; Keisuke Horikawa; Debra L. Dugger; Wendy Sandoval; Susmith Mukund; Anuradha Zindal; Flavius Martin; Clifford Quan; Jeffrey Tom; Wayne J. Fairbrother; Michael J. Townsend; Søren Warming; Jason DeVoss; Jinfeng Liu; Erin C. Dueber; Patrick Caplazi; Wyne P. Lee; Christopher C. Goodnow

Inactivation of the TNFAIP3 gene, encoding the A20 protein, is associated with critical inflammatory diseases including multiple sclerosis, rheumatoid arthritis and Crohn’s disease. However, the role of A20 in attenuating inflammatory signalling is unclear owing to paradoxical in vitro and in vivo findings. Here we utilize genetically engineered mice bearing mutations in the A20 ovarian tumour (OTU)-type deubiquitinase domain or in the zinc finger-4 (ZnF4) ubiquitin-binding motif to investigate these discrepancies. We find that phosphorylation of A20 promotes cleavage of Lys63-linked polyubiquitin chains by the OTU domain and enhances ZnF4-mediated substrate ubiquitination. Additionally, levels of linear ubiquitination dictate whether A20-deficient cells die in response to tumour necrosis factor. Mechanistically, linear ubiquitin chains preserve the architecture of the TNFR1 signalling complex by blocking A20-mediated disassembly of Lys63-linked polyubiquitin scaffolds. Collectively, our studies reveal molecular mechanisms whereby A20 deubiquitinase activity and ubiquitin binding, linear ubiquitination, and cellular kinases cooperate to regulate inflammation and cell death.


Journal of Biological Chemistry | 2012

Furin-cleaved Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) Is Active and Modulates Low Density Lipoprotein Receptor and Serum Cholesterol Levels

Michael T. Lipari; Wei Li; Paul Moran; Monica Kong-Beltran; Tao Sai; Joyce Lai; S. Jack Lin; Ganesh Kolumam; Jose Zavala-Solorio; Anita Izrael-Tomasevic; David Arnott; Jianyong Wang; Andrew S. Peterson; Daniel Kirchhofer

Background: Two forms of PCSK9, an intact and a furin cleaved form, circulate in blood. Results: Both forms, as highly purified recombinant proteins, are able to bind to and trigger degradation of LDL receptors and elevate serum cholesterol levels. Conclusion: Furin cleavage is not associated with a loss of PCSK9 polypeptides or a significant loss of function. Significance: LDL-c levels are controlled by both forms of PCSK9. Proprotein convertase subtilisin/kexin 9 (PCSK9) regulates plasma LDL cholesterol levels by regulating the degradation of LDL receptors. Another proprotein convertase, furin, cleaves PCSK9 at Arg218-Gln219 in the surface-exposed “218 loop.” This cleaved form circulates in blood along with the intact form, albeit at lower concentrations. To gain a better understanding of how cleavage affects PCSK9 function, we produced recombinant furin-cleaved PCSK9 using antibody Ab-3D5, which binds the intact but not the cleaved 218 loop. Using Ab-3D5, we also produced highly purified hepsin-cleaved PCSK9. Hepsin cleaves PCSK9 at Arg218-Gln219 more efficiently than furin but also cleaves at Arg215-Phe216. Further analysis by size exclusion chromatography and mass spectrometry indicated that furin and hepsin produced an internal cleavage in the 218 loop without the loss of the N-terminal segment (Ser153–Arg218), which remained attached to the catalytic domain. Both furin- and hepsin-cleaved PCSK9 bound to LDL receptor with only 2-fold reduced affinity compared with intact PCSK9. Moreover, they reduced LDL receptor levels in HepG2 cells and in mouse liver with only moderately lower activity than intact PCSK9, consistent with the binding data. Single injection into mice of furin-cleaved PCSK9 resulted in significantly increased serum cholesterol levels, approaching the increase by intact PCSK9. These findings indicate that circulating furin-cleaved PCSK9 is able to regulate LDL receptor and serum cholesterol levels, although somewhat less efficiently than intact PCSK9. Therapeutic anti-PCSK9 approaches that neutralize both forms should be the most effective in preserving LDL receptors and in lowering plasma LDL cholesterol.


Cell Reports | 2015

Glucagon Couples Hepatic Amino Acid Catabolism to mTOR-Dependent Regulation of α-Cell Mass

Mark Solloway; Azadeh Madjidi; Chunyan Gu; Jeff Eastham-Anderson; Holly J. Clarke; Noelyn M. Kljavin; Jose Zavala-Solorio; Lance Kates; Brad A. Friedman; Matt Brauer; Jianyong Wang; Oliver Fiehn; Ganesh Kolumam; Howard M. Stern; John B. Lowe; Andrew S. Peterson; Bernard B. Allan

Understanding the regulation of islet cell mass has important implications for the discovery of regenerative therapies for diabetes. The liver plays a central role in metabolism and the regulation of endocrine cell number, but liver-derived factors that regulate α-cell and β-cell mass remain unidentified. We propose a nutrient-sensing circuit between liver and pancreas in which glucagon-dependent control of hepatic amino acid metabolism regulates α-cell mass. We found that glucagon receptor inhibition reduced hepatic amino acid catabolism, increased serum amino acids, and induced α-cell proliferation in an mTOR-dependent manner. In addition, mTOR inhibition blocked amino-acid-dependent α-cell replication ex vivo and enabled conversion of α-cells into β-like cells in vivo. Serum amino acids and α-cell proliferation were increased in neonatal mice but fell throughout postnatal development in a glucagon-dependent manner. These data reveal that amino acids act as sensors of glucagon signaling and can function as growth factors that increase α-cell proliferation.


EBioMedicine | 2015

Sustained Brown Fat Stimulation and Insulin Sensitization by a Humanized Bispecific Antibody Agonist for Fibroblast Growth Factor Receptor 1/βKlotho Complex

Ganesh Kolumam; Mark Z. Chen; Raymond K. Tong; Jose Zavala-Solorio; Lance Kates; Nicholas van Bruggen; Jed Ross; Shelby K. Wyatt; Vineela D. Gandham; Richard A. D. Carano; Diana Ronai Dunshee; Ai-Luen Wu; Benjamin Haley; Keith R. Anderson; Søren Warming; Xin Y. Rairdan; Nicholas Lewin-Koh; Yingnan Zhang; Johnny Gutierrez; Amos Baruch; Thomas Gelzleichter; Dale Stevens; Sharmila Rajan; Travis W. Bainbridge; Jean-Michel Vernes; Y. Gloria Meng; James Ziai; Robert Soriano; Matthew J. Brauer; Yongmei Chen

Dissipating excess calories as heat through therapeutic stimulation of brown adipose tissues (BAT) has been proposed as a potential treatment for obesity-linked disorders. Here, we describe the generation of a humanized effector-less bispecific antibody that activates fibroblast growth factor receptor (FGFR) 1/βKlotho complex, a common receptor for FGF21 and FGF19. Using this molecule, we show that antibody-mediated activation of FGFR1/βKlotho complex in mice induces sustained energy expenditure in BAT, browning of white adipose tissue, weight loss, and improvements in obesity-associated metabolic derangements including insulin resistance, hyperglycemia, dyslipidemia and hepatosteatosis. In mice and cynomolgus monkeys, FGFR1/βKlotho activation increased serum high-molecular-weight adiponectin, which appears to contribute over time by enhancing the amplitude of the metabolic benefits. At the same time, insulin sensitization by FGFR1/βKlotho activation occurs even before the onset of weight loss in a manner that is independent of adiponectin. Together, selective activation of FGFR1/βKlotho complex with a long acting therapeutic antibody represents an attractive approach for the treatment of type 2 diabetes and other obesity-linked disorders through enhanced energy expenditure, insulin sensitization and induction of high-molecular-weight adiponectin.


PLOS ONE | 2013

Antibody-Mediated Activation of FGFR1 Induces FGF23 Production and Hypophosphatemia

Ai-Luen Wu; Bo Feng; Mark Z. Chen; Ganesh Kolumam; Jose Zavala-Solorio; Shelby K. Wyatt; Vineela D. Gandham; Richard A. D. Carano; Junichiro Sonoda

The phosphaturic hormone Fibroblast Growth Factor 23 (FGF23) controls phosphate homeostasis by regulating renal expression of sodium-dependent phosphate co-transporters and cytochrome P450 enzymes involved in vitamin D catabolism. Multiple FGF Receptors (FGFRs) can act as receptors for FGF23 when bound by the co-receptor Klotho expressed in the renal tubular epithelium. FGFRs also regulate skeletal FGF23 secretion; ectopic FGFR activation is implicated in genetic conditions associated with FGF23 overproduction and hypophosphatemia. The identity of FGFRs that mediate the activity of FGF23 or that regulate skeletal FGF23 secretion remains ill defined. Here we report that pharmacological activation of FGFR1 with monoclonal anti-FGFR1 antibodies (R1MAb) in adult mice is sufficient to cause an elevation in serum FGF23 and mild hypophosphatemia. In cultured rat calvariae osteoblasts, R1MAb induces FGF23 mRNA expression and FGF23 protein secretion into the culture medium. In a cultured kidney epithelial cell line, R1MAb acts as a functional FGF23 mimetic and activates the FGF23 program. siRNA-mediated Fgfr1 knockdown induced the opposite effects. Taken together, our work reveals the central role of FGFR1 in the regulation of FGF23 production and signal transduction, and has implications in the pathogenesis of FGF23-related hypophosphatemic disorders.


Cytokine | 2010

Lymphotoxin-αβ heterotrimers are cleaved by metalloproteinases and contribute to synovitis in rheumatoid arthritis

Judy Young; Xin Yu; Kristen Wolslegel; Allen Nguyen; Catherine Kung; Eugene Chiang; Ganesh Kolumam; Nathan Wei; Wai Lee Wong; Laura DeForge; Michael J. Townsend; Jane L. Grogan

Tumor necrosis factor-superfamily (TNF-SF) members, lymphotoxin (LT)-alpha and LTbeta, are proinflammatory cytokines associated with pathology in rheumatoid arthritis. LTalpha3 homotrimers are secreted, whereas LTalpha(1)beta(2) heterotrimers are expressed on the surface of activated lymphocytes. As many TNF-SF members are actively cleaved from cell membranes, we determined whether LTalphabeta heterotrimers are also cleaved, and are biologically active in rheumatoid arthritis (RA) patients. LTalphabeta heterotrimers were detected in culture supernatants from activated human T-helper (Th) 0, Th1, and Th17 cells, together with LTalpha3 and TNFalpha. The heterotimers were actively cleaved from the cell surface by ADAM17 metalloproteinase (MMP) and MMP-8, and cleavage was inhibited by TAPI-1, a TNF-alpha converting enzyme (TACE) inhibitor. Soluble LTalphabeta was detected in serum from both normal donors and RA patients, and was elevated in synovial fluid from RA patients compared to osteoarthritis (OA) patients. Levels of LTalphabeta in RA patient synovial fluid correlated with increased TNFalpha, IL-8, IL-12, IL-1beta, IFN-gamma, and IL-6 cytokines. Moreover, recombinant LTalpha1beta2-induced CXCL1, CXCL2, IL-6, IL-8, VCAM-1, and ICAM-1 from primary synovial fibroblasts isolated from RA patients. Therefore, soluble LTalphabeta in synovial fluid is associated with a proinflammatory cytokine milieu that contributes to synovitis in RA.

Collaboration


Dive into the Ganesh Kolumam's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge