The invention relates to compounds and methods (including methods of treatment) to restore normal metabolic function in humans, including but not limited to normal glucose levels. The invention in one embodiment contemplates methods of reducing elevated glucose levels in subjects with elevated glucose levels by administering a composition comprising at least a portion of human Sfrp5. The invention in one embodiment contemplates methods of reducing elevated glucose levels in subjects with elevated glucose levels by administering a composition comprising an inhibitor of Wnt5a, including but not limited to an antibody inhibitor. In addition, the present invention contemplates compounds and methods for detecting and measuring metabolic function.
Obesity is a major health problem that is linked to the development of metabolic disorders that are associated with a low-grade inflammatory state in adipose tissue. It is increasingly recognized that adipose tissue secretes a variety of bioactive substances that are referred to as adipokines [1-3]. The majority of these adipokines are pro-inflammatory including TNFα, IL-6 and leptin. However, the well-studied cytokine adiponectin is anti-inflammatory and it promotes insulin-sensitization and cardio-protection [2, 4]. However, adiponectin cannot be practically used because the amounts needed for therapeutic treatment are too great.
Clearly, other safe and effective treatments are needed for human administration.
The invention relates to compositions and methods (including methods of treatment) to restore normal metabolic function in humans, including but not limited to normal glucose levels. The invention in one embodiment contemplates methods of reducing elevated glucose levels in subjects with elevated glucose levels by administering a composition comprising at least a portion of human Sfrp5 (or a nucleic acid construct capable of expressing human Sfrp5). The invention in one embodiment contemplates methods of reducing elevated glucose levels in subjects with elevated glucose levels by administering a composition comprising an inhibitor of Wnt5a, including but not limited to an antibody inhibitor, such as a humanized antibody with affinity for Wnt5a. In addition, the present invention contemplates compounds and methods for detecting and measuring metabolic function.
In one embodiment the invention relates to a host cell comprising an expression vector, said vector encoding human Sfrp5 or a portion thereof. In further embodiments, said host cell is capable of expressing said human Sfrp5 or portion thereof as a soluble protein at a level greater than or equal to 5% of the total cellular protein. In further embodiments, said host cell is capable of expressing said human Sfrp5 or portion thereof as a soluble protein at a level greater than or equal to 15% of the total cellular protein. In further embodiments, said vector encodes a portion consisting of a domain of human Sfrp5. In further embodiments, said vector encodes a fusion protein comprising at least a portion of human Sfrp5, said portion comprising a portion of the sequence of SEQ ID NO: 2. In further embodiments, said fusion protein comprises a poly-histidine tract. In further embodiments, said fusion protein comprises at least a portion of an immunoglobulin molecule. In further embodiments, said portion of an immunoglobulin molecule consists of an Fc fragment.
In another embodiment the invention relates to a soluble fusion protein comprising at least a portion of human Sfrp5, said portion comprising a portion of the sequence of SEQ ID NO: 2. In another embodiment the invention relates to the fusion protein, wherein said portion consists of at least one domain of human Sfrp5. In another embodiment the invention relates to the fusion protein, wherein said fusion protein comprises a poly-histidine tract. In another embodiment the invention relates to the fusion protein, wherein said fusion protein comprises at least a portion of an immunoglobulin molecule. In another embodiment the invention relates to the fusion protein, wherein said portion of an immunoglobulin molecule consists of an Fc fragment. In another embodiment the invention relates to the fusion protein, wherein said fusion protein is substantially endotoxin-free.
In another embodiment the invention relates to a method of reducing elevated glucose levels, comprising: a) providing a subject with elevated glucose levels and a composition comprising at least a portion of human Sfrp5; b) administering said composition to said subject; and c) measuring said glucose levels of said subject until they are reduced. In another embodiment the invention relates to a method of reducing elevated glucose levels, wherein said subject is a human. In another embodiment relates to a method, wherein said elevated glucose levels are reduced by at least 20%. In another embodiment relates to a method, wherein said elevated glucose levels are reduced by at least 40%. In another embodiment relates to a method, wherein said composition comprises a fusion protein comprising at least a portion of human Sfrp5, said portion comprising a portion of the sequence of SEQ ID NO: 2. In another embodiment relates to a method, wherein said portion consists of at least one domain of human Sfrp5. In another embodiment relates to a method, wherein said fusion protein comprises a poly-histidine tract. In another embodiment relates to a method, wherein said fusion protein comprises at least a portion of an immunoglobulin molecule. In another embodiment relates to a method, wherein said portion of an immunoglobulin molecule consists of an Fc fragment.
In another embodiment the invention relates to a method of reducing elevated glucose levels, comprising: a) providing a subject with elevated glucose levels and a composition comprising an expression vector, said vector capable of expressing at least a portion of human Sfrp5 in vivo; b) administering said composition to said subject; and c) measuring said glucose levels of said subject until they are reduced. In another embodiment the invention relates to a method of reducing elevated glucose levels, comprising: a) providing a subject with elevated glucose levels and an implantable device, said device capable of releasing at least a portion of human Sfrp5 in vivo; b) implanting said device in said subject; and c) measuring said glucose levels of said subject until they are reduced.
In another embodiment the invention relates to a method of reducing elevated glucose levels, comprising: a) providing a subject with elevated glucose levels and a composition comprising an antibody (e.g. humanized antibody) or portion thereof reactive with human Wnt5a; b) administering said composition to said subject; and c) measuring said glucose levels of said subject until they are reduced. The present invention also contemplates a composition comprising humanized monoclonal antibody reactive with human Wnt5a.
In another embodiment, the present invention contemplates a method of measuring metabolic function (by measuring markers in tissue or blood, preferably plasma or serum), comprising: providing i) a sample (e.g. tissue, blood, secretion, etc.) from a subject and ii) a reagent (or other means) for measuring human Sfrp5 protein (or fragments thereof) or human Sfrp5 nucleic acid (or portions thereof); measuring the level of Sfrp5 protein or nucleic acid as an indicator of metabolic function. In one embodiment, said reagent is an antibody reactive with human Sfrp5 protein. In another embodiment, said antibody is specific for human Sfrp5 (i.e. not reactive with other human proteins). In another embodiment, said antibody is reactive with human, but unreactive with mouse Sfrp5. The present invention contemplates these antibodies as compositions. In one embodiment, said reagent is an oligonucleotide probe (with a region of complementarity for human Sfrp5 nucleic acid) for measuring human Sfrp5 mRNA. In one embodiment, mRNA is measured in tissue biopsies.
As used herein, the term “fusion protein” refers to a chimeric protein containing the protein of interest (i.e., human Sfrp5 or fragments thereof) joined to an exogenous protein fragment (the fusion partner which consists of another protein or protein fragment). The fusion partner may enhance solubility or half-life of the Sfrp5 protein or protein fragment as expressed in a (preferably human) host cell, and may also provide an affinity tag to allow purification of the recombinant fusion protein from the host cell or culture supernatant, or both. If desired, the fusion protein may be removed from the protein of interest prior to administration by a variety of enzymatic or chemical means known to the art.
As used herein, the term “poly-histidine tract” when used in reference to a fusion protein refers to the presence of two to ten histidine residues at either the amino- or carboxy-terminus of a protein of interest, i.e. Sfrp5 or portion thereof (e.g. a domain). A poly-histidine tract of six to ten residues is preferred. The poly-histidine tract is also defined functionally as being a number of consecutive histidine residues added to the protein of interest which allows the affinity purification of the resulting fusion protein on a nickel-chelate column.
The term “subject” includes humans and non-human animals. In the case of humans, the term includes both in-patients and out-patients, and particularly the elderly and the obese, whether or not under the care of a medical professional.
As used herein “immunoglobulin” refers to any of a group of large glycoproteins that are secreted by plasma cells and that function as antibodies in the immune response by binding with specific antigens. The specific antigen bound by an immunoglobulin may or may not be known. There are five classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM.
The twin “antibody,” as used herein, is intended to refer to immunoglobulin molecules comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains (lambda or kappa) inter-connected by disulfide bonds. An antibody has a known specific antigen with which it binds. Each heavy chain of an antibody is comprised of a heavy chain variable region (abbreviated herein as HCVR, HV or VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL or KV or LV to designate kappa or lambda light chains) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR). Each variable region (VH or VL) contains 3 CDRs, designated CDR1, CDR2 and CDR3. Each variable region also contains 4 framework sub-regions, designated FR1, FR2, FR3 and FR4.
As used herein, the term “antibody fragments” refers to a portion of an intact antibody. Examples of antibody fragments include, but are not limited to, linear antibodies, single-chain antibody molecules, Fv, Fab and F(ab′)2 fragments, and multispecific antibodies formed from antibody fragments. The antibody fragments preferably retain at least part of the heavy and/or light chain variable region.
As used herein, “humanized” forms of non-human (e.g., murine) antibodies are antibodies that contain minimal (e.g. less tan 10%) sequence, or no sequence, derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity. In some instances, Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are generally made to further refine antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a nonhuman immunoglobulin and all or substantially all of the FR residues are those of a human immunoglobulin sequence. The humanized antibody may also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. Examples of methods used to generate humanized antibodies are described in U.S. Pat. No. 5,225,539 to Winter et al. (herein incorporated by reference) [5].
Importantly, early methods for humanizing antibodies often resulted in antibodies with lower affinity than the non-human antibody starting material. More recent approaches to humanizing antibodies address this problem by making changes to the CDRs. See U.S. Patent Application Publication No. 20040162413, hereby incorporated by reference. In some embodiments, the present invention provides an optimized heteromeric variable region (e.g. that may or may not be part of a full antibody other molecule) having equal or higher antigen binding affinity than a donor heteromeric variable region, wherein the donor heteromeric variable region comprises three light chain donor CDRs, and wherein the optimized heteromeric variable region comprises: a) a light chain altered variable region comprising; i) four unvaried human germline light chain framework regions, and ii) three light chain altered variable region CDRs, wherein at least one of the three light chain altered variable region CDRs is a light chain donor CDR variant, and wherein the light chain donor CDR variant comprises a different amino acid at only one, two, three or four positions compared to one of the three light chain donor CDRs (e.g. the at least one light chain donor CDR variant is identical to one of the light chain donor CDRs except for one, two, three or four amino acid differences).
The binding “affinity” is directly related to the ratio of the off-rate constant (generally reported in units of inverse time, e.g., seconds.sup.-1) to the on-rate constant (generally reported in units of concentration per unit time, e.g., molar/second). The binding affinity may be determined by, for example, an ELISA assay, kinetic exclusion assay or surface plasmon resonance. Qualitatively, an antibody with affinity for a protein such as Wnt5a demonstrates higher levels of binding as compared to binding to other proteins.
Sfrp5 is predicted to be a secreted protein based upon the presence of a signal peptide and the absence of a transmembrane domain. Sfrp5 is a member of the Sfrp family that contains a cysteine-rich domain homologous to the putative Wnt-binding site of Frizzled proteins. This family of protein acts as soluble modulators that sequesters Wnt proteins in the extracellular space between cells and prevents their binding to the receptors and antagonizes Wnt-mediated signaling pathways [6, 7], and canonical Wnt signaling negatively regulates adipogenesis [8]. However, to date, nothing is known about the role of Sfrp5 in regulation of obesity-associated metabolism on the control of non-canonical Wnt signaling in adipose tissue.
Ideally, recombinant proteins are expressed as soluble proteins at high levels (i.e., greater than or equal to about 0.75% of total cellular protein, and more preferably, greater than 5% or even 15% of total cellular protein) in host cells. This facilitates the production and isolation of sufficient quantities in a highly purified form (i.e., substantially free of endotoxin or other pyrogen contamination).
In one embodiment, the present invention contemplates expressing and producing human Sfrp5 or fragment thereof as a fusion protein. In one embodiment, the fusion protein comprises a poly-histidine tract (also called a histidine tag). In one embodiment, the fusion protein comprises a portion of an antibody, e.g. the Fc fragment. The production of fusion proteins is not limited to the use of a particular expression vector and host strain. Several commercially available expression vectors and host strains can be used to express the C fragment protein sequences as a fusion protein containing a histidine tract. For example, Qiagen has a pQE xpression vector for mammalian cells.
In another embodiment, the present invention contemplates a method of measuring metabolic function (by measuring markers in tissue or blood, preferably plasma or serum), comprising: providing i) a sample (e.g. tissue, blood, secretion, etc.) from a subject and ii) a reagent (or other means) for measuring human Sfrp5 protein (or fragments thereof) or human Sfrp5 nucleic acid (or portions thereof); measuring the level of Sfrp5 protein or nucleic acid in the sample as an indicator of metabolic function.
In one embodiment, said reagent is an antibody reactive with human Sfrp5 protein. It is important to stress that commercially available antibodies that are advertised as reactive with human Sfrp5 protein were tested and found as not reactive or effective. Therefore, an antibody is contemplated against a unique human Sfrp5 sequence. In one embodiment, a polyclonal antibody against human Sfrp5 is generated by immunizing rabbits (or a monoclonal antibody is generated by immunizing mice or rats) with one or more synthetic peptides reflecting a unique portion of the human Sfrp sequence (such as WAPARCEEYDYYGWQAEP; SEQ ID NO: 5); in one embodiment, one or more peptides of this type are conjugated to KLH (e.g. through the Cys via maleimide linkage). In a preferred embodiment, said antibody is specific for human Sfrp5 (i.e. not reactive with other human proteins). In another embodiment, said antibody is reactive with human, but unreactive with mouse Sfrp5. The present invention contemplates these antibodies as compositions. In one embodiment, said reagent is an oligonucleotide probe (with a region of complementarity for human Sfrp5 nucleic acid) for measuring human Sfrp5 mRNA. In one embodiment, mRNA is measured in tissue biopsies.
A variety of administration approaches (e.g. introducing an expression vector into a subject) and routes of administration may be used. However, it is preferred that administration of the recombinant protein (or fragment thereof) be done intravenously or through an implantable device that provides Sfrp5 in therapeutic quantities.
Because the dysregulation of adipokines can contribute to the pathophysiology of various obesity-linked disorders, we sought to identify new adipokine candidates by performing microarray analysis on the adipose tissues of lean and high-fat/high-sucrose (HF/HS) diet-induced obese mice [9]. The transcript encoding secreted frizzled-related protein (Sfrp) 5 was significantly higher in epididymal adipose tissue in obese mice fed HF/HS diet for 12 weeks than in lean mice fed a normal diet (
Sfrp5 is predicted to be a secreted protein based upon the presence of a signal peptide and the absence of a transmembrane domain using signal IP and SOUSI software, respectively. Sfrp5 is a member of the Sfrp family that contains a cysteine-rich domain homologous to the putative Wnt-binding site of Frizzled proteins. Without limiting the invention in any manner to any particular mechanism, it is believed that this family of protein acts as soluble modulators that sequesters Wnt proteins in the extracellular space between cells and prevents their binding to the receptors and antagonizes Wnt-mediated signaling pathways [6, 7], and canonical Writ signaling negatively regulates adipogenesis [8]. However, to date, nothing is known about the role of Sfrp5 in regulation of obesity-associated metabolism on the control of non-canonical Wnt signaling in adipose tissue.
Sfrp5 has been shown to bind and antagonize both Wnt5a and Wnt11 [10]. Furthermore, non-canonical Wnt pathway is activated by Wnt5a class ligands including Wnt5a and Wnt11 [11]. Thus, we assessed protein expression of Wnt5a and Wnt11 in epididymal fat tissues in WT mice fed normal or HF/HS diet by immunoblot analysis. Little or no expression of Wnt5a was observed in adipose tissues of normal diet-fed mice, but its level was increased in adipose tissue of HF/HS diet-fed obese mice after 12 and 24 weeks of HF/HS diet feeding (
Because obesity-related metabolic dysfunction is attributed to inflammation and oxidative and ER stress in adipose tissue [12-16], the expression of TNFα, NADPH oxidase components gp91phox and P47phox, macrophage markers (F4/80 and CD68) and markers of ER stress (GRP78 and CHOP) was assessed in epididymal fat pads in lean mice and compared to that in obese mice fed HF/HS diet for 12 or 24 weeks. Corresponding to the increase in Wnt5a/Sfrp5 expression ratio, adipose expression of TNFα, gp91phox, P47phox, F4/80, CD68, GRP78 and CHOP was significantly higher in mice fed HF/HS diet for 12 weeks than in lean mice, and the levels of these transcripts were further elevated in mice fed HF/HS diet for 24 weeks (
The visceral fat of obese individuals was scored for the presence of macrophage crown-like structures (CLS), an indicator of adipose tissue inflammation [17, 18]. CLS-positive individuals exhibited a decrease in Sfrp5 transcript expression compared with obese individuals that were negative for CLS (
Sfrp5 expression by cultured 3T3-L1 adipocytes was up-regulated when cells were induced to differentiate, with the highest level of expression attained by day 4 (
Sfrp5-deficient (Sfrp5−/−) mice in a C57BL/6 background were used to investigate the pathophysiological role of Sfrp5 in the regulation of metabolism and adipose tissue inflammation. Sfrp5−/− mice were fertile and viable. Despite the absence of Sfrp5 protein expression in adipose tissue (
Histological analyses were performed on epididymal adipose tissues. Sfrp5−/− mice fed a HF/HS diet had adipocytes with larger cross-section area than HF/HS diet fed WT mice (
An increased inflammatory response in adipose tissues is linked to the development of insulin resistance and glucose intolerance [14-16]. Thus, macrophage content in the adipose tissue of Sfrp5−/− and WT mice was assessed. Cells positive for F4/80, a macrophage marker, appeared at a significantly higher frequency in adipose tissue of Sfrp5−/− mice compared to WT mice when both strains were fed a HF/HS diet ((
The pathways involved in canonical or non-canonical Wnt signaling were assessed in epididymal adipose tissues of Sfrp5−/− and WT mice fed a HF/HS diet. No differences were detected in transcript expression of cyclin D1 or WISP2, indicators of canonical Wnt signaling, between Sfrp5−/− and WT mice (
To assess the effect of Sfrp5 and Wnt5a on JNK activation at the cellular level, 3T3-L1 adipocytes were transduced with adenoviral vectors expressing Sfrp5 (Ad-Sfrp5) or β-galactosidase (Ad-β-gal) as control, followed by incubation with Wnt5a protein. Transduction of 3T3-L1 cells with Ad-Sfrp5 led to an increase in Sfrp5 protein in both cell lysates and media compared with Ad-β-gal (
To assess the effect of Sfrp5 on JNK activation and inflammatory response in macrophages in vitro, cultured murine macrophages were stimulated with Wnt5a protein in the presence of conditioned media from 3T3-L1 adipocytes transduced with Ad-Sfrp5 or Ad-β-gal. Wnt5a-treatment stimulated JNK phosphorylation in macrophages in conditioned media from Ad-β-gal-treated 3T3-L1 adipocytes, which was blocked by the conditioned media from Ad-Sfrp5-transduced adipocytes (
Mice lacking both Sfrp5 and Jnk1 were generated to investigate the causal role of JNK1 activation in the severe diet-induced metabolic dysfunction and adipose tissue inflammation that develops in the Sfrp5-deficient mice. Consistent with a previous report [20], Jnk1−/− mice exhibited improvements in insulin resistance and glucose intolerance, and reduced BW compared with WT mice when fed the HF/HS diet (
To test whether the over-expression of Sfrp5 is protective against the development of insulin resistance and glucose intolerance in vivo, we intravenously administered Ad-Sfrp5 or Ad-β-gal to WT and Sfrp5−/− mice that were fed a HF/HS diet for 10 weeks. Detectable circulating levels of Sfrp5 could be measured in serum one week after the delivery of Ad-Sfrp5 (
The development of obesity-related metabolic disorders is attributed in part to an imbalance in the production of adipokines, most of which are pro-inflammatory and detrimental to metabolism. In comparison, adiponectin is unique among adipokines in that it has anti-inflammatory and insulin-sensitizing actions in a number of models. Here we show that Sfrp5 is secreted by adipocytes and that it regulates the microenvironment of white adipose tissue by regulating macrophage activation under conditions of metabolic stress. Whereas Sfrp5−/− mice do not express a detectable phenotype when fed a normal chow diet, these animals displayed aggravated fat pad inflammation and systemic metabolic dysfunction when fed a high calorie diet. Conversely, the acute administration of Sfrp5 to models of obese and diabetic mice improved metabolic function and reduced adipose tissue inflammation. Notably, the salutary actions of Sfrp5 administration on glucose metabolism were more effective when compared with the acute over-production of adiponectin in ob/ob mice.
Our data indicate that Sfrp5 can function to neutralize noncanonical JNK activation by Wnt5a in macrophages and adipocytes via paracrine and autocrine mechanisms, respectively (
As noted above, one approach to treatment involves administering human Sfrp5 protein to a subject. However, because of the findings described above, another approach to treatment is contemplated, i.e. administering antibodies to Wnt5a. Thus, in one embodiment, the present invention contemplates a method of reducing elevated glucose levels, comprising: a) providing a subject with elevated glucose levels and a composition comprising an antibody or portion thereof reactive with human Wnt5a; b) administering said composition to said subject; and measuring said glucose levels of said subject until they are reduced. It is preferred that said subject is a human and said antibody is a humanized monoclonal antibody reactive with human Wnt5a.
It is not intended that the present invention be limited to a particular antibody of method of making an anti-Wnt5a antibody.
In one embodiment, the present invention contemplates the use of an implant or implantable device to provide either human Sfrp5 protein or portion thereof (directly as a protein or via an expression vector, including a vector in host cells) or antibody to Wnt5a. There are a variety of devices available and it is not intended that the present invention be limited to a particular device. For example, there are devices comprising tubular matrices (U.S. Pat. No. 6,716,225, hereby incorporated by reference [25]) which can be applied in this manner. In one embodiment, the implant comprises polymeric gel material containing bioactive molecules (see U.S. Pat. No. 6,290,729, hereby incorporated by reference [26]). In one embodiment, the implant comprises a sponge-like structure having a plurality of convoluted capillaries and from which the active material is released (see U.S. Pat. No. 4,587,267, hereby incorporated by reference [27]).
Phospho-Akt (Ser473), phospho-JNK (Thr183/Tyr185), phospho-cJUN (Ser63), Akt, and GAPDH antibodies were purchased from Cell Signaling Technology. Phospho-IRS-1 (Ser307) antibody was purchased from Upstate biotechnology. Tunicamycin, H2O2 and β-actin antibody were purchased from Sigma Chemical Co. The polyclonal antibody against mouse Sfrp5 was generated by immunizing rabbits with two synthetic peptides conjugated to KLH through the Cys via maleimide (APTRGQEYDYYGWQAEP: amino acid residues 22-38 (SEQ ID NO: 7) and Acetyl-VKMRIKEIKIDNGDRKLIG: amino acid residues 201-219) (21st Century Biochemicals) (SEQ ID NO: 8). SP600125 was purchased from Biomol international. Recombinant mouse Wnt5a protein produced in CHO cells (endotoxin free: <1.0 EU/μg protein by the LAL method), mouse Wnt5a antibody and mouse TNFα proteins were purchased from R&D Systems.
Mice lacking Sfrp5 were backcrossed and maintained on the C57BL6/J background. Sfrp5−/− mice were generated by replacing the first protein coding exon with the PGKneobpAloxA cassette as described previously [28]. Sfrp5−/− mice and littermate wild-type (WT) C57BL6/J mice were used. To generate mice lacking both Sfrp5 and JNK1, Sfrp5−/− mice and Jnk1−/+ mice (Jackson laboratory) were inbred. Ob/ob mice were purchased from Jackson laboratory. Study protocols were approved by the Boston University Institutional Animal Care and Use Committee. Mice were fed either a normal chow diet (Harlan Teklad global 18% protein rodent diet, #2018) or a HF/HS diet (Bio-Serv, #F1850) [9] as indicated. The composition of the HF/HS diet was 35.8% fat (primarily lard), 36.8% carbohydrate (primarily sucrose), and 20.3% protein. For the high caloric diet feeding, mice at the age of 10 weeks were maintained on a HF/HS diet for 12 or 24 weeks.
Visceral adipose tissue was collected during gastric bypass surgery in obese human adults (age ≥21 years) with a body mass index ≥30 kg/m2 as described previously [17]. Patients with unstable medical conditions such as active coronary syndromes, congestive heart failure, systemic infection, malignancy, or pregnancy were excluded. The presence or absence of macrophage crown-like structures (CLS) in adipose tissue was determined by immunohistochemical stains with CD68 in a blinded manner as described previously [17]. The insulin resistance marker, homeostasis model assessment of insulin resistance (HOMA-IR) were quantified from blood samples [17]. All subjects gave written, informed consent, and the study was approved by the Boston University Medical Center Institutional Review Board.
Mouse 3T3-L1 cells (ATCC) were maintained in DMEM with 10% fetal bovine serum (FBS) and differentiated into adipocytes by treatment with DMEM supplemented with 5 μg/ml of insulin, 0.5 mM 1-methyl-3-isobutyl-xanthin, and 1 μM dexamethazone [29]. At day 7 after differentiation, 3T3-L1 adipocytes were treated with tunicamycin, H2O2, TNFα or vehicle for 24 h. Peritoneal macrophages from lean WT C57BL/6 mice were maintained in DMEM supplemented 10% FBS and placed in DMEM with 0.5% FBS for 16 h for serum starvation. Macrophages were incubated in the conditioned media from 3T3-L1 adipocytes transduced with adenoviral vectors in the presence of Wnt5a protein (200 ng/ml) or vehicle for 24 h. In some experiments, cells were pretreated with SP600125 (15 μM) or vehicle for 1 h followed by treatment with Wnt5a protein.
Construction of Adenoviral Vectors
For transduction experiments, tetracycline-regulated adenovirus (Ad) vectors were used [30]. The Ad vectors expressing β-galactosidase (β-gal) or Sfrp5 were constructed under the control of seven consecutive tetracycline-responsive elements (TRE) and a CMV minimal promoter (AdTRE-β-gal or AdTRE-Sfrp5) Co-transfection of AdTRE vector with Ad vectors encoding tetracycline transactivator (tTA, a fusion of TRE-binding protein and VP16 transactivation domain) under the control of the CMV promoter/enhancer (AdCMV-tTA) results in activation of transgene. Ad vectors expressing adiponectin (Ad-APN) under control of the CMV promoter was constructed as previously described [31, 32]. For in vivo gene transfer, AdTRE-β-gal or AdTRE-Sfrp5 along with AdCMV-tTA (2.5×108 pfu for each adenovirus) was injected into the jugular vein of mice. For some experiments, Ad-APN (5.0×108 pfu total) was intravenously administered to mice. For in vitro transduction, cells were transfected with AdTRE-β-gal or AdTRE-Sfrp5 in the presence of AdCMV-tTA (125 MOI for each adenovirus) for 24 h. The media was then replaced with fresh DMEM, and cells were incubated for additional 24 h. Cells were treated with or without recombinant Wnt5a protein (200 ng/ml) for indicated length of time. For detection of Sfrp5 in media, protein was concentrated 3-fold with a Microcon column.
3T3-L1 adipocytes were transduced with adenoviral vectors for 24 h and co-transfected with a TOPflash (Upstate) construct and a Renilla luciferase control plasmid (pRL-SV40, Promega) to normalize for transfection efficiency. After transfection, cells were treated with Wnt5a protein (200 ng/ml) or vehicle. Cells were lysed and analyzed on a luminometer by using dual luciferase assay kit (Promega).
Serum insulin levels were determined by ELISA using mouse insulin as a standard (Crystal Chemical Inc.). Serum glucose, free fatty acid and triglyceride levels were measured with enzymatic kits (Wako Chemicals). Glucose tolerance testing was performed on 6 hr-fasted mice injected intraperitoneally with D-glucose (1 g/kg body weight) [9]. Blood glucose levels were determined immediately before, and 30, 60, 90, and 120 min after injection as determined by an Accu-Chek glucose monitor (Roche Diagnostics Corp.). Insulin tolerance testing was performed on 6 hr-fasted mice injected intraperitoneally with human insulin (Humulin R, Eli Lilly) at 1.5 U/kg body weight for lean mice or at 4.5 U/kg body weight for obese mice. Blood glucose levels were determined immediately before and 15, 30, and 60 min after injection. Insulin signaling in adipose tissues was determined by measurement of Akt phosphorylation following insulin administration. Mice were fasted overnight and treated with insulin (4.5 U/kg body weight) or saline via the inferior vena cava. Epididymal fat was isolated after 4 min, and immunoblot analysis was preformed. To determine triglyceride content of liver, lipid was extracted using the Bligh-Dyer method [33]. Liver tissues were homogenized in chloroform/MeOH/H2O (1:2:0.8) and centrifuged. Supernatants were collected, and equal amounts of chloroform and H2O were added. After vortexing and centrifugation, the chloroform layer was collected, dried completely and resuspended in isopropanol containing 10% Triton-X. Triglyceride levels were measured with enzymatic kits (Wako Chemicals).
The sections of epididymal adipose tissue were fixed in 10% formalin, dehydrated, and embedded in paraffin. Adipose tissue sections were stained with hematoxylin and eosin to examine the morphology and with anti-F4/80 antibody (Santa Cruz) to detect macrophages. Adipocyte cross-sectional areas were measured in 200 cells per mouse using Image J software. Macrophage accumulation was quantified by measuring the number of F4/80-positive cells per mm2 in 20 randomly chosen microscopic fields. Liver tissues were embedded in OCT compound (Sakura Finetech USA Inc.) and snap frozen in liquid nitrogen and stained with oil red O for lipid deposition by standard methods [9].
Epididymal fat pads from male wild-type C57BL/6 mice fed a normal diet were excised, minced in PBS and digested with 1 mg/ml collagenase Type 1 (Worthington Chemical Corporation) at 37° C. for 30 min. The digested fat tissue was filtered through a mesh and centrifuged at 1000 rpm for 5 min to separate floating adipocytes from the SV fraction.
Measurement of mRNA Levels
Gene expression level was quantified by real-time PCR. Total RNA was prepared with the use of a Qiagen kit. cDNA was produced using ThermoScript RT-PCR Systems (Invitrogen). PCR was performed on iCycler iQ Real-Time PCR Detection System (Bio-Rad) using SYBR Green 1 as a double standard DNA specific dye (Applied Biosystems).
Tissue and cell samples were homogenized, and equal amounts of proteins were separated with denaturing SDS 4-15% polyacrylamide gels. Following transfer to membranes, immunoblot analysis was performed with the indicated antibodies followed by incubation with secondary antibody conjugated with horseradish peroxidase at a 1:5000 dilution. ECL Western Blotting Detection kit (Amersham Pharmacia Biotech) or ECL Advance Western Blotting Detection kit (Amersham Pharmacia Biotech) was used for detection. Relative phosphorylation or protein levels were quantified by Image J program.
All data are expressed as means±SEM. Differences were analyzed by Student's unpaired t test or analysis of variance (ANOVA) for multiple comparisons. A value of P<0.05 was accepted as statistically significant.
Number | Date | Country | |
---|---|---|---|
61332051 | May 2010 | US |
Number | Date | Country | |
---|---|---|---|
Parent | 13100895 | May 2011 | US |
Child | 15267712 | US |
Number | Date | Country | |
---|---|---|---|
Parent | 15267712 | Sep 2016 | US |
Child | 16259150 | US |